Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR THE TREATMENT OF FAT INFILTRATION IN MUSCLE
Document Type and Number:
WIPO Patent Application WO/2019/246225
Kind Code:
A1
Abstract:
This disclosure provides methods of using compositions comprising amino acid entities to reduce fat infiltration in muscle, particularly under conditions of muscle atrophy. The disclosure also provides methods for enhancing muscle function by reducing fat infiltration in the muscle comprising administering an effective amount of the compositions to a subject in need thereof.

Inventors:
CHAKRAVARTHY MANU (US)
COMB WILLIAM (US)
HAMILL MICHAEL (US)
TRAMONTIN TONY (US)
Application Number:
PCT/US2019/037936
Publication Date:
December 26, 2019
Filing Date:
June 19, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AXCELLA HEALTH INC (US)
International Classes:
A61K31/198; A61K31/4172; A61K38/00; A61P21/00; G01N24/00
Domestic Patent References:
WO2017202939A12017-11-30
WO2017085138A12017-05-26
WO2019036471A12019-02-21
Foreign References:
CN101049500A2007-10-10
US20170196944A12017-07-13
US20140343112A12014-11-20
US20180207119A12018-07-26
US20180207119A12018-07-26
US201715847289A2017-12-19
Other References:
ODESSA ADDISON ET AL: "Intermuscular Fat: A Review of the Consequences and Causes", INTERNATIONAL JOURNAL OF ENDOCRINOLOGY, vol. 2014, 1 January 2014 (2014-01-01), US, pages 1 - 11, XP055621774, ISSN: 1687-8337, DOI: 10.1155/2014/309570
TANYA M. HOLLOWAY ET AL: "A Novel Amino Acid Composition Ameliorates Short-Term Muscle Disuse Atrophy in Healthy Young Men", FRONTIERS IN NUTRITION, vol. 6, 1 January 2019 (2019-01-01), pages 105, XP055622142, DOI: 10.3389/fnut.2019.00105
EVANS ET AL., JOURNAL OF CACHEXIA, SARCOPENIA AND MUSCLE, vol. 10, 2019, pages 14 - 21
ADDISON, INT. J. ENDOCRINOLOGY, 2014, Retrieved from the Internet
BHANJI ET AL., HEP. INTL., 2018, Retrieved from the Internet
MALIETZIS ET AL., ANN SURG., vol. 263, no. 2, February 2016 (2016-02-01), pages 320 - 5
SUEDA ET AL., DIS COLON RECTUM., vol. 61, no. 3, March 2018 (2018-03-01), pages 364 - 374
MELIS ET AL., ORTHOPAEDICS & TRAMATOLOGY: SURGERY & RESEARCH, vol. 95, 2009, pages 319 - 324
Attorney, Agent or Firm:
COLLAZO, Diana M. et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method for reducing fat infiltration in muscle comprising administering to a subject at risk of fat infiltration in muscle a composition comprising:

a) a leucine amino acid entity, an arginine amino acid entity, and a glutamine amino acid entity;

b) a N-acetylcysteine (NAC) entity; and

c) an essential amino acid (EAA)-entity chosen from a histidine amino acid-entity, a lysine amino acid-entity, a phenylalanine amino acid-entity, and a threonine amino acid-entity or a combination of two, three, or four of the EAAs.

2. The method of claim 1, wherein the subject at risk of fat infiltration in muscle has a rotator cuff injury.

3. The method of claim 2, wherein administration of the composition precedes a surgery for the rotator cuff injury.

4. The method of claim 2, further comprising determining a level of fat infiltration in shoulder muscle affected by the rotator cuff injury before surgery.

5. The method of claim 2, further comprising determining a level of fat infiltration in shoulder muscle affected by the rotator cuff injury after surgery.

6. The method of claim 2, wherein the subject is an elderly subject.

7. The method of claim 1, wherein the subject at risk of fat infiltration in muscle has chronic back pain (fat infiltration in paraspinal muscles); HIV patients (fat infiltration in locomotor muscles); spinal cord injury; stroke; COPD; end-stage liver disease (ESLD), e.g., hepatic encephalopathy, variceal bleeding, portal hypertension, ascites, infection risk, sepsis, all- cause hospitalization, and all-cause and liver-related mortality; and muscle weakness associated with ageing.

8. The method of claim 1, wherein administering the composition to the subject at risk of fat infiltration in muscle improves a muscle function of sequestering glucose.

9. The method of claim 8, wherein the subject at risk for fat infiltration in muscle has diabetes or metabolic disease.

10. The method of claim 1, wherein the subject at risk of fat infiltration in muscle has cancer.

11. The method of claim 10, wherein the cancer is colorectal cancer or periampullary cancer.

12. The method of claim 1, wherein the subject at risk for fat infiltration in muscle does not have significant increase in BMI, sarcopenia, or other overt conditions.

13. The method of claim 1, wherein the subject at risk for fat infiltration in muscle suffers from cirrhosis without sarcopenia.

14. The method of claim 1, wherein the subject at risk for fat infiltration in muscle has ESLD, e.g., hepatic encephalopathy, variceal bleeding, portal hypertension, ascites, infection risk, sepsis, all-cause hospitalization, and all-cause and liver-related mortality.

15. The method of claim 1, wherein the degree of fat infiltration in muscle is determined by magnetic resonance imaging (MRI).

16. The method of claim 1, wherein at least one of methionine (M), trytophan (W), valine (V), or cysteine (C) is absent, or if present, is present at less than 1 weight (wt.) %.

17. The composition of claim 1, wherein the total wt. % of (a)-(c) is greater than the total wt. % of any other amino acid entity in the composition.

18. The method of any of the preceding claims, wherein the composition further comprises an isoleucine amino acid entity, a valine amino acid entity, or both an isoleucine amino acid entity and a valine amino acid entity.

19. The method of any of the preceding claims, wherein at least one of the leucine amino acid entity, the arginine amino acid, the glutamine amino acid entity, or one, two, three, or all of the EAA amino acid entities is a free amino acid, and at least 50 wt. % of the total dry wt. of the composition is one or more amino acid entities in free form.

20. The method of any of the preceding claims, wherein at least one of the leucine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, or one, two, three, or all of the EAA amino acid entities is in salt form., and at least 50 wt. % of the total dry wt. of the composition is one or more amino acid entities in salt form.

21. The method of any of the preceeding claims, wherein the composition comprises about 0.5 g to about 15 g of the leucine amino acid entity, about 0.25 g to about 10 g of the isoleucine amino acid entity, about 0.25 g to about 10 g of the valine amino acid entity, about 0.5 to about 25 g of the arginine amino acid entity, about 0.5 g to about 20 g of the glutamine amino acid entity, about 0.1 to about 5 g the NAC or a salt thereof, about 0.05 g to about 3 g of the L- histidine or a salt thereof, about 0.05 to about 6 g of the L-lysine or a salt thereof, about 0.04 to about 2 g of the L-phenylalanine or a salt thereof, and about 0.08 to about 4 g of the L-threonine or a salt thereof entity; e.g., about 1 g of the leucine amino acid entity, about 0.5 g of the isoleucine amino acid entity, about 0.5 g of the valine amino acid entity, about 1.5 g or about 1.81 of the arginine amino acid entity, about 1.33 g of the glutamine amino acid entity, about 0.15 g or about 0.3 g of the NAC or a salt thereof, about 0.08 g of the L-histidine or a salt thereof, about 0.35 g of the L-lysine or a salt thereof, about 0.08 g of the L-phenylalanine or a salt thereof, and about 0.17 g of the L-threonine or a salt thereof.

22. The method of any of the preceeding claims wherein the composition is a pharmaceutical composition and further comprises a pharmaceutically acceptable excipient.

23. A method for improving muscle function in reduced mobility or immobilized muscle by reducing fat infiltration, wherein the method comprises administering to a subject in need thereof an effective amount of a composition comprising at least four amino acids, wherein the composition reduces fat infiltration in muscle.

24. The method of claim 23, wherein fat infiltration is reduced in the reduced-mobility or immobilized muscle.

25. The method of claim 23, further comprising determining a degree of fat infiltration in muscle.

26. The method of claim 23, wherein the composition comprises:

a) a leucine amino acid entity, an arginine amino acid entity, and a glutamine amino acid entity;

b) a N-acetylcysteine (NAC) entity, e.g., NAC; and

c) an essential amino acid (EAA)-entity chosen from a histidine amino acid-entity, a lysine amino acid-entity, a phenylalanine amino acid-entity, and a threonine amino acid-entity or a combination of two, three, or four of the EAAs;

provided at:

d) at least one amino acid entity is not provided as a peptide of more than 20 amino acid residues in length; and wherein:

(i) the amino acid entity of (a) is selected from Table 1; and

(ii) one or both of the arginine amino acid entity and the glutamine amino acid entity are present at a higher amount (wt. %) than the leucine amino acid entity.

27. The method of any of claims 23-26, wherein the subject has a disease or disorder selected from the group consisting of a rare muscle disease, muscle atrophy, sarcopenia, muscle deterioration, muscle decay, cachexia, drug-induced myopathy, muscular dystrophy, myopenia, muscle weakness, perceived muscle weakness, ICU-acquired myopathy, bums-related myopathy, a neuromuscular disorder, ventilator-induced diaphragmatic dystrophy, hyponatremia, hypokalemia, a calcium deficiency, hypercalcemia, amyotrophic lateral sclerosis, a bone weakness disease; or the subject has or is identified as having decreased muscle function due to aging, injury, muscle atrophy, infection, disease, stroke, or a fracture or other trauma, and wherin the fracture or other trauma is selected from rotator cuff surgery, knee surgery, hip surgery, joint replacement, injury repair surgery, or has worn a cast.

28. The method of any one of claims 27, which comprises evaluating fat infiltration in muscle in the subject to evaluate effectiveness of administration of the composition in treating the disease or disorder, decreased muscle function, or fracture or other trauma.

29. A method for determining whether a composition comprising at least four amino acids is effective in treating a disease or disorder associated with muscle function comprising administering to the subject a composition comprising at least four amino acids and determining whether there is a reduction in fat infiltration in muscle in the subject.

30. The method of claim 29, wherein fat infiltration is in muscle tissue affected by the disease or disorder associated with muscle function.

Description:
COMPOSITIONS AND METHODS

FOR THE TREATMENT OF FAT INFILTRATION IN MUSCLE

BACKGROUND

There are many diseases and disorders associated with increased infiltration of fat in muscle, e.g., muscle atrophy. Muscle atrophy is associated with cancer, AIDS, renal failure, liver disease, spinal cord injury, and congestive heart failure. Furthermore, disuse of muscles through immobilization or aging also results in muscle atrophy with increased fat infusion in muscle.

The gold standard for treating muscle atrophy conditions is recovery of function.

However, direct assessment of muscle functional muscle mass is challenging (Evans et al., 2019, Journal of Cachexia, Sarcopenia and Muscle, 10:14-21). Furthermore, the ability to reduce infiltration of fat in muscle is an important therapeutic target, with few if any pharmacological agents available.

Thus, there is a need to identify pharmacological methods of reducing fat infiltration in muscle, and to use such agents to reduce fat infiltration in muscle, particularly under conditions of muscle atrophy. Furthermore, there is a need to identify markers of functional muscle mass to develop additional metabolic rebalancing compositions for enhancing muscle mass and function (relative to not receiving any therapy), such as for treating muscle-related disease and disorders where fat infiltration in muscle occurs.

SUMMARY

The invention provides method for reducing fat infiltration in muscle comprising administering to a subject at risk of fat infiltration in muscle a composition comprising an Active Moiety. The Active Moiety comprises:

a) a leucine amino acid entity, an arginine amino acid entity, and a glutamine amino acid entity;

b) a N-acetylcysteine (NAC) entity; and

c) an essential amino acid (EAA)-entity chosen from a histidine amino acid-entity, a lysine amino acid-entity, a phenylalanine amino acid-entity, and a threonine amino acid-entity or a combination of two, three, or four of the EAAs. The method can be used to reduce fat infiltration in muscle of a subject at risk of fat infiltration in muscle who has a rotator cuff injury, and in particular administration of the composition precedes a surgery for the rotator cuff injury. The invention can include determining a level of fat infiltration in shoulder muscle affected by the rotator cuff injury, e.g., before surgery or after surgery. In a specific embodiment, the subject with a rotator cuff injury is an elderly subject.

Alternatively, the method can be used to reduce fat infiltration in muscle of a subject at risk of fat infiltration in muscle who has chronic back pain (fat infiltration in paraspinal muscles); HIV patients (fat infiltration in locomotor muscles); spinal cord injury; stroke; COPD; end-stage liver disease (ESLD), e.g., hepatic encephalopathy, variceal bleeding, portal hypertension, ascites, infection risk, sepsis, all-cause hospitalization, and all-cause and liver- related mortality; and muscle weakness associated with ageing. Futhermore, the administering the composition to the subject at risk of fat infiltration in muscle can improve a muscle function of sequestering glucose, e.g., when the subject at risk for fat infiltration in muscle has diabetes or metabolic disease.

In yet another alternative, the subject at risk of fat infiltration in muscle has cancer, e.g., colorectal cancer or periampullary cancer. Preferably the cancer is treated surgically in conjuction with the methods of the invention.

In still another alternative, the subject at risk for fat infiltration in muscle does not have significant increase in BMI, sarcopenia, or other overt conditions, or the subject at risk for fat infiltration in muscle suffers from cirrhosis without sarcopenia.

Very few reliable methods are available to measure intramuscular fat infiltration (IMF); magnetic resonance imaging (MRI) is one of them. Computer tomography (CT) can also be used, but it is not as efficient at MRI.

A significant advantage of the invention is the ability to finely regulate the amount and relative ratio of each amino acid in the composition, which is not possible with peptides of more than 20 amino acids in length, including proteins. Thus, in the methods of the invention, the composition preferably does not include a peptide of more than 20 amino acid residues in length. In another aspect, at least one of methionine (M), trytophan (W), valine (V), or cysteine (C) is absent, or if present, is present at less than 1 weight (wt.) % of dry weight, particulary dry weight of the Active Moiety. Furthermore, the composition can further comprise an isoleucine amino acid entity, a valine amino acid entity, or both an isoleucine amino acid entity and a valine amino acid entity.

While the methods envision compositions of amino acid entities, in specific embodiments exemplified in the application, at least one of the leucine amino acid entity, the arginine amino acid, the glutamine amino acid entity, or one, two, three, or all of the EAA amino acid entities is a free amino acid; all of them can be free amino acids. Thus, it is possible that at least 50 wt. % of the total dry wt. of the composition is one or more amino acid entities in free form.

Alternatively, at least one of the leucine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, or one, two, three, or all of the EAA amino acid entities is in salt form; all of them can be in salt form. Thus, it is possible that at least 50 wt. % of the total dry wt. of the composition is one or more amino acid entities in salt form.

As demonstrated in the example, the method can be practiced with a composition comprising about 0.5 g to about 15 g of the leucine amino acid entity, about 0.25 g to about 10 g of the isoleucine amino acid entity, about 0.25 g to about 10 g of the valine amino acid entity, about 0.5 to about 25 g of the arginine amino acid entity, about 0.5 g to about 20 g of the glutamine amino acid entity, about 0.1 to about 5 g the NAC or a salt thereof, about 0.05 g to about 3 g of the L-histidine or a salt thereof, about 0.05 to about 6 g of the L-lysine or a salt thereof, about 0.04 to about 2 g of the L-phenylalanine or a salt thereof, and about 0.08 to about 4 g of the L-threonine or a salt thereof entity; e.g., about 1 g of the leucine amino acid entity, about 0.5 g of the isoleucine amino acid entity, about 0.5 g of the valine amino acid entity, about 1.5 g or about 1.81 of the arginine amino acid entity, about 1.33 g of the glutamine amino acid entity, about 0.15 g or about 0.3 g of the NAC or a salt thereof, about 0.08 g of the L-histidine or a salt thereof, about 0.35 g of the L-lysine or a salt thereof, about 0.08 g of the L-phenylalanine or a salt thereof, and about 0.17 g of the L-threonine or a salt thereof.

According to the invention, any method can be practices with a composition that is a pharmaceutical composition. Thus, the composition can further comprise a pharmaceutically acceptable excipient, such as an excipient that is suitable for oral administration.

More broadly, the invention includes a method for improving muscle function in reduced-mobility or immobilized muscle by reducing fat infiltration, wherein the method comprises administering to a subject in need thereof an effective amount of a composition comprising at least four amino acids, wherein the composition reduces fat infiltration in muscle, e.g., in the reduced-mobility or immobilized muscle. This method can be practiced with a composition that is a pharmaceutical composition. Thus, the composition can further comprise a pharmaceutically acceptable excipient, such as an excipient that is suitable for oral

administration. The discovery that such compositions are capable of reducing infiltration of fat in muscle thus allows for determining a degree of fat infiltration in muscle under conditions of reduced-mobility or immobilized muscle. Any method for determining or evaluating the degree of fat infiltration in muscle can be used, e.g., MRI, DEXA, or CT.

In these foregoing method for improving muscle function in reduced-mobility or immobilized muscle by reducing fat infiltration, wherein the composition can comprise:

a) a leucine amino acid entity, an arginine amino acid entity, and a glutamine amino acid entity;

b) a N-acetylcysteine (NAC) entity, e.g., NAC; and

c) an essential amino acid (EAA)-entity chosen from a histidine (H)-amino acid-entity, a lysine (K)-amino acid-entity, a phenylalanine (F)-amino acid-entity, and a threonine (T)-amino acid-entity or a combination of two, three, or four of the EAAs. Furthermore:

d) at least one amino acid entity is not provided as a peptide of more than 20 amino acid residues in length. In still another aspect:

(i) the amino acid entity of (a) is selected from Table 1; and

(ii) one or both of the arginine amino acid entity and the glutamine amino acid entity are present at a higher amount (wt. %) than the leucine amino acid entity.

In these foregoing method for improving muscle function in reduced-mobility or immobilized muscle by reducing fat infiltration, the subject can have a disease or disorder selected from the group consisting of a rare muscle disease, muscle atrophy, sarcopenia, muscle deterioration, muscle decay, cachexia, drug-induced myopathy, muscular dystrophy, myopenia, muscle weakness, perceived muscle weakness, ICET-acquired myopathy, bums-related myopathy, a neuromuscular disorder, ventilator-induced diaphragmatic dystrophy, hyponatremia, hypokalemia, a calcium deficiency, hypercalcemia, amyotrophic lateral sclerosis, and a bone weakness disease. Alternatively, the subject can have or be identified as having decreased muscle function due to aging, injury, muscle atrophy, infection, disease, stroke, or a fracture or other trauma. The fracture or other trauma may be selected from rotator cuff surgery, knee surgery, hip surgery, joint replacement, injury repair surgery, or the subject has worn a cast. In the case of a fracture or trauma, the subject can receive the composition after the fracture or other trauma or before the fracture or other trauma, in the latter case, e.g., in conjunction with planned elective surgery. For example, the subject may have a rotator cuff injury, and further the subject may have rotator cuff surgery. The invention also provides for determining or evaluating fat infiltration in muscle in the subject to evaluate effectiveness of administration of the composition in treating the disease or disorder, decreased muscle function, or fracture or other trauma, e.g., prior to an elective procedure. According to the invention and exemplified below, determining or evaluating fat infiltration in muscle reveals that a fat fraction in muscle is unchanged from before the treatment, or even improved. As noted above, any method for determining or evaluating the degree of fat infiltration in muscle can be used, e.g., MRI, DEXA, or CT.

In yet another embodiment, the invention provides method for determining whether a composition comprising at least four amino acids is effective in treating a disease or disorder associated with muscle function. This method comprises administering to the subject a composition comprising at least four amino acids and determining whether there is a reduction in fat infiltration in muscle in the subject. Thus, fat infiltration in muscle can serve as a surrogate for muscle function in a study, e.g., a clinical trial, post-marketing trial, prognostic assay, etc. in conjunction with treatment with a composition comprising at least four amino acids. In particular, fat infiltration can be in muscle tissue affected by the disease or disorder associated with muscle function.

Thus, in the situation where the subject has has a rotator cuff injury, the method can be used. For example, administration of the composition can precede a surgery for the rotator cuff injury. In this situation, the invention provides for determining a level of fat infiltration in shoulder muscle affected by the rotator cuff injury before surgery. In conjunction with evaluating progression and prognosis, it is also possible to determine a level of fat infiltration in shoulder muscle affected by the rotator cuff injury after surgery.

In the method for determining whether a composition comprising at least four amino acids is effective in treating a disease or disorder associated with muscle function, the subject at risk of fat infiltration in muscle has chronic back pain (fat infiltration in paraspinal muscles);

HIV patients (fat infiltration in locomotor muscles); spinal cord injury; stroke; COPD; ESLD, e.g., hepatic encephalopathy, variceal bleeding, portal hypertension, ascites, infection risk, sepsis, all-cause hospitalization, and all-cause and liver-related mortality; and muscle weakness associated with ageing. Alternatively, the method improves a muscle function of sequestering glucose in a subject at risk of fat infiltration in muscle, e.g., if the subject has diabetes or metabolic disease. In yet another alternative, the subject at risk of fat infiltration in muscle has cancer, such as colorectal cancer or periampullary cancer. Moreover, the subject at risk for fat infiltration in muscle may not have significant increase in BMI, sarcopenia, or other overt conditions. Thus, the subject at risk for fat infiltration in muscle may suffer from cirrhosis without sarcopenia, or may have ESLD, e.g., hepatic encephalopathy, variceal bleeding, portal hypertension, ascites, infection risk, sepsis, all-cause hospitalization, and all-cause and liver- related mortality. As noted above, any method for determining or evaluating the degree of fat infiltration in muscle can be used, e.g., MRI, DEXA, or CT.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG 1. Change in vastus lateralis cross-sectional area (CSA) and fiber types by histology during limb immobilization (Day 15 vs. Day 8).

FIGS 2A and 2B. Change in quadriceps fat fraction during immobilization (Day 15 vs. Day 8) in both immobilized and nonimmobilized legs. 2A. Representative images depicting fat fraction (FF) changes in the immobilized and non-immobilized thighs of a subject who received treatment and a subject who received placebo. Purple represents fat fraction; red represents muscle fraction. 2B. Plot of fat fraction changes quantified across all subjects.

FIG 3. Change in vastus lateralis electrical impedance measurement (EIM) parameters (phase, max reactance, and change in reactance slope) during immobilization & recovery. Data are mean ± SEM from n = 9 (Pbo) and n = 10 (therapeutic).

DETAILED DESCRIPTION

The present invention is based on the unprecedented discovery that compositions comprising amino acid entities are capable of reducing fat infiltration in muscle. This discovery provides for treatment of a number of diseases and disorders involving fat infiltration in muscle. In some cases, fat infiltration in muscle results in increased morbidity, worsening of the disease or disorder, or predicts a worse outcome from an intervention such as surgery to repair a torn rotator cuff.

Thus the invention provides, at least in part, methods of reducing fat infiltration in muscle by administering an Active Moiety composition of the invention, which is a composition comprising at least four different amino acid entities. In some cases, the invention further comprises determining or evaluating the extent or degree of infiltration of fat in muscle, e.g., as a diagnostic, a prognostic indicator, to evaluate progress of the disease or disorder with or without treatment, or as a surrogate of muscle health, or any combination of the foregoing. Various techniques are available for determining the extent of infiltration of fat in muscle. The most rigorous techniques are computed tomograph (CT) and magnetic resonance imaging (MRI).

The methods of reducing fat infiltration in muscle may also provide a method of treating any or all of immobilization, malnutrition, fasting, aging, autophagy, reduced protein synthesis, anabolic resistance, junction integrity (e.g., neuromuscular junction integrity), insulin resistance, decreased mitochondrial biogenesis, an energy deficit, or anaplerosis in a subject that includes administering to a subject in need thereof an effective amount of a pharmaceutical composition including defined amino acid components. In some embodiments, the subject has a rare muscle disease. In some embodiments, the subject has sarcopenia, muscle deterioration, decay, atrophy, cachexia, drug-induced myopathy, muscular dystrophy, or myopenia. In some embodiments, the subject has a fracture or other trauma. In some embodiments, the subject has a drug-induced myopathy. In some embodiments, the subject has a statin-induced myopathy. In some embodiments, the subject has a steroid-induced myopathy. In some embodiments, the subject has an immunosuppressant-induced myopathy. In some embodiments, the subject has a

chemotherapeutic-induced myopathy. In some embodiments, the subject has an alcohol-induced myopathy.

In addition to evaluating or determining the extent of fat infiltration in muscle, improvements in muscle function can be assessed by performing metrics selected from maximal isometric knee strength test (e.g., to determine changes in muscle strength), muscle biopsy (e.g., to determine muscle fiber quality), and electrical impedance myography (EIM) (e.g., to determine muscle health, such as resistive and capacitive properties of muscle tissue and sensitivity to disuse-related atrophy), or other standard clinical performance assessments such as the Short Performance Physical Battery (SPPB), Harris Hip Score and others. In some embodiments, the composition is for use as a medicament in improving muscle function in a subject at risk of or experiencing fat infiltration in muscle. In some embodiments, the composition is for use as a medicament in treating a muscle disease or disorder involving fat infiltration in the muscle in a subject.

In some embodiments, the composition is for use in the manufacture of a medicament for improving muscle function in a subject at risk of or experiencing fat infiltration in muscle.

In some embodiments, the composition is for use in the manufacture of a medicament for treating a muscle disease or disorder involving fat infiltration in the muscle in a subject.

Additionally, the composition may be useful as a dietary supplement, e.g., a nutritional supplement, dietary formulation, functional food, medical food, food, or beverage comprising a composition described herein. Another embodiment provides a nutritional supplement, dietary formulation, functional food, medical food, food, or beverage comprising a composition described herein for use in the management of any of the diseases or disorders described herein.

One embodiment provides a method of maintaining or improving muscle health, muscle function, muscle functional performance, or muscle strength, comprising administering to a subject an effective amount of a composition described herein to reduce fat infiltration in muscle in the subject. Another embodiment provides a method of providing nutritional support or supplementation to a subject suffering from muscle atrophy comprising administering to the subject an effective amount of a composition described herein to reduce fat infiltration in muscle in the subject. Yet another embodiment provides a method of providing nutritional support or supplementation that aids in the management of muscle atrophy to a subject comprising administering to the subject in need thereof an effective amount of a composition described herein to reduce fat infiltration in muscle in the subject.

Definitions

Terms used in the claims and specification are defined as set forth below unless otherwise specified.

It must be noted that, as used in the specification and the appended claims, the singular forms“a,”“an” and“the” include plural referents unless the context clearly dictates otherwise. The term“fat infiltration in muscle” means an increase in the fat fraction of muscle below the muscle fascia, as distinguished from subcutaneous fat. For example, fat found in the deep fascia of the thigh is fat infiltration in muscle. Various publications use terms such as intramuscular fat fraction, intermuscular fat infiltration, intramuscular fat, intermuscular fat, intermuscular adipose tissue (IMAT), and myosteatosis or myostasis. All of these terms refer to fat (visible storage of lipids in adipocytes) located between muscle fibers and between muscle groups, or within myocytes themselves. Fat that has infiltrated in muscle appears to have some of the same characteristic as ectopic or visceral fat, e.g., fat in liver or other organs, or in the abdomen.

“Reduction of fat infiltration in muscle” means that the degree or extent of fat infiltration in a subject, e.g., to muscle that is immobilized, injured, or otherwise subject to muscle infiltration, is less than it would have been in the absence of an intervention, i.e., in the absence of administering an Active Moiety to the subject. Thus, the fat fraction in the muscle is lower than it would have otherwise been. In one example, administration of an Active Moiety reduces fat infiltration in muscle by 100%., i.e., it prevents it. However, the invention includes any reduction in fat infiltration that would occur in the absence of treatment with an Active Moiety. Preferably reduction of fat infiltration in muscle is detectable. More preferably it is significant, e.g., reaches statistical significance in a population of subjects in a controlled study.

The term“fat fraction” or“FF” refers to the fraction of fat or percentage of fat in a limb, part of a limb, or body (taking the entire volume of the limb, part of a limb, or body as the whole or 100%). Similarly, the muscle mass is the fraction of muscle or percentage of muscle in limb, part of a limb, or body.

In the Example described below, images depicting fat fraction (FF) show that in a subject administered placebo, the subject’s non-immobilized leg had no change in FF, while the subject’s immobilized leg had increased FF and decreased muscle mass. By contrast, in a subject administered an Active Moiety, the immobilized leg had lower fat fraction and a higher muscle content following immobilization compared to placebo. These FF changes were quantified across all subjects. Percent change in quadriceps muscle fat fraction increased more that 10% in subjects who received placebo, compared to about 0% in subjects who received Active Moiety.

The term“subject” refers to a human person, and can include, but is not limited to, a patient, i.e., a person who is under care of a healthcare provider (doctor, nurse practitioner, etc.). It can also mean a person in a clinical study, or a person who self-diagnoses and self-treats, or a subject who receives a dietary supplement (as broadly defined above).

As used herein, the term“Active Moiety” means a combination of four or more amino acid entities that, in aggregate, have a physiological effect. In some cases, the physiological effect can be a therapeutic effect involving reduction of fat infiltration into muscle, as defined below. For example, an Active Moiety can rebalance a metabolic dysfunction in a subject suffering from a disease or disorder. An Active Moiety of the invention can contain other biologically active ingredients. In some examples, the Active Moiety comprises a defined combination of four or more amino acid entities, as set out in detail below. The individual amino acid entities are present in the Active Moiety in various amounts or ratios, which can be presented as amount by weight (e.g., in grams), ratio by weight of amino acid moieties to each other, amount by mole, amount by dry weight percent of the Active Moiety, amount by mole percent of the Active Moiety, caloric content, percent caloric contribution to the Active Moiety, etc. Generally, this disclosure will provide grams of amino acid entity in a dosage form, weight percent of an amino acid moiety relative to the weight of the Active Moiety, i.e., the weight of all the amino acid moieties and any other biologically active ingredient present in the Active Moiety, or in ratios. The abbreviation“wt.” means weight.

As used herein, the term“amino acid entity” refers to an amino acid in one or both of free form or salt form, an amino acid residue of a peptide (e.g., of a dipeptide, tripeptide, or polypeptide of 20 amino acids or less in length), a derivative of an amino acid, a precursor of an amino acid, or a metabolite of an amino acid (see, e.g., Table 1). Accordingly, the term“XXX amino acid entity” refers to an amino acid entity that if a free amino acid, comprises free XXX or XXX in salt form; if a peptide, refers to a peptide comprising an XXX residue; if a derivative, refers to a derivative of XXX; if a precursor, refers to a precursor of XXX; and if a metabolite, refers to a XXX metabolite. For example, where XXX is leucine, then leucine amino acid entity refers to free leucine or leucine in salt form, a peptide of less than 20 amino acids comprising a leucine residue, a leucine derivative, a leucine precursor, or a metabolite of leucine (where such derivative, precursor, or metabolite achieves the same physiological effect as leucine); where XXX is arginine, then arginine amino acid entity refers to free arginine or arginine in salt form, a peptide of less than 20 amino acids comprising an arginine residue, an arginine derivative, an arginine precursor, or a metabolite of arginine (where such derivative, precursor, or metabolite achieves the same physiological effect as arginine); where XXX is glutamine, then glutamine amino acid entity refers to free glutamine or glutamine in salt form, a peptide of less than 20 amino acids comprising a glutamine residue, a glutamine derivative, a glutamine precursor, or a metabolite of glutamine (where such derivative, precursor, or metabolite achieves the same physiological effect as glutamine); where XXX is N-acetylcysteine (NAC), then NAC-amino acid entity refers to free NAC or NAC in salt form, a peptide comprising a NAC residue, a NAC derivative, a NAC precursor, or a metabolite of NAC (where such derivative, precursor, or metabolite achieves the same physiological effect as NAC); where XXX is histidine (H), then histidine amino acid entity refers to free histidine or histidine in salt form, a peptide of less than 20 amino acids comprising a histidine residue, a histidine derivative, a histidine precursor, or a metabolite of histidine (where such derivative, precursor, or metabolite achieves the same physiological effect as histidine); where XXX is lysine, then lysine amino acid entity refers to free lysine or lysine in salt form, a peptide of less than 20 amino acids comprising comprising a lysine residue, a lysine derivative, a lysine precursor, or a metabolite of lysine (where such derivative, precursor, or metabolite achieves the same physiological effect as lysine); where XXX is phenylalanine, then phenylalanine amino acid entity refers to free phenylalanine or phenylalanine in salt form, a peptide of less than 20 amino acids comprising comprising a phenylalanine residue, a phenylalanine derivative, a phenylalanine precursor, or a metabolite of phenylalanine (where such derivative, precursor, or metabolite achieves the same physiological effect as phenylalanine); or where XXX is threonine, then threonine amino acid entity refers to free threonine or threonine in salt form, a peptide of less than 20 amino acids comprising a threonine residue, a threonine derivative, a threonine precursor, or a metabolite of threonine (where such derivative, precursor, or metabolite achieves the same physiological effect as threonine). Where the biological system provides for isomerization of a D-amino acid to the L- form, the D-amino acid can be an amino acid entity.

Salts of amino acids include any physiologically tolerable, e.g., ingestible, salt. For pharmaceutical compositions, the salt form of an amino acid present in the Active Moiety should be a pharmaceutically acceptable salt. In a specific example, the salt form is the hydrochloride (HC1) salt form of the amino acid. In some embodiments, the derivative of an amino acid entity comprises an amino acid ester (e.g., an alkyl ester, e.g., an ethyl ester or a methyl ester of an amino acid entity) or a keto- acid. Table 1. Amino acid entities include amino acids, precursors, metabolites, and derivatives of the compositions described herein.

“About” and“approximately” shall generally mean an acceptable degree of error for the quantity measured given the nature or precision of the measurements. Exemplary degrees of error are within 20 percent (%), typically, within 10%, and more typically, within 5% of a given value or range of values.

An“amino acid” refers to an organic compound having an amino group (-NH 2 ), a carboxylic acid group (-C(=0)0H), and a side chain bonded through a central carbon atom, and includes essential and non- amino acids, as well as natural and unnatural amino acids.

The proteogenic amino acids, shown below, are known by three- and one-letter abbreviations in addition to their full names. For a given amino acid, these abbreviations are used interchangeably herein. For example, Feu, F or leucine all refer to the amino acid leucine; Ile, I or isoleucine all refer to the amino acid isoleucine; Val, V or valine all refer to the amino acid valine; Arg, R or arginine all refer to the amino acid arginine; and Gln, Q or glutamine all refer to the amino acid glutamine. Fikewise, the non-natural amino acid derivative N- acetylcysteine may be referred to interchangeably by“NAC” or“N-acetylcysteine.” Amino acids may be present as F-isomers of amino acids to ensure physiological activity.

Table 2. Canonical (proteogenic) amino acid names and abbreviations

A“branched chain amino acid” is an amino acid selected from leucine, isoleucine, and valine.

The term“effective amount” as used herein means an amount of an Active Moiety, or pharmaceutical composition comprising an Active Moiety, which is sufficient enough to significantly and positively modify the symptoms and/or conditions to be treated (e.g., provide a positive clinical response). The effective amount of an active ingredient for use in a

pharmaceutical composition will vary with the particular condition being treated, the severity of the condition, the duration of treatment, the nature of concurrent therapy, the particular active ingredient(s) being employed, the particular pharmaceutically-acceptable excipient(s) and/or carrier(s) utilized, and like factors with the knowledge and expertise of the attending physician.

A“pharmaceutical composition” described herein comprises at least one amino acid and a pharmaceutically acceptable carrier or excipient. In some embodiments, the pharmaceutical composition is used as a therapeutic, a nutraceutical, a medical food, or as a supplement.

The term“pharmaceutically acceptable” as used herein, refers to amino acids, materials, excipients, compositions and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

A“therapeutic effect” means a beneficial clinical effect. A beneficial clinical effect can be shown by lessening the progression of a disease and/or alleviating one or more symptoms of the disease. Preferably, the beneficial clinical effect is statistically significant. A“health effect” in the context of a food supplement means that there is some benefit to the normal structure or function of a human or target organ in a human.

A“unit dose” or“unit dosage” as used herein means an amount or dose of medicine prepared in an individual packet or container for convenience, safety, or monitoring. A“unit dose” or“unit dosage” comprises the drug product or drug products in the form in which they are marketed for use, with a specific mixture of active ingredients and inactive components

(excipients), in a particular configuration (such as a capsule shell, for example), and apportioned into a particular dose.

As used herein, the terms“treat,”“treating,” or“treatment” refer in one embodiment, to ameliorating a disease or disorder of fat infiltration in muscle (i.e., slowing or arresting or reducing the development of the disease or disorder or at least one of the clinical symptoms thereof). In another embodiment,“treat,”“treating,” or“treatment” refers to alleviating or ameliorating at least one physical parameter of fat infiltration in muscle, including those which may not be discernible by the patient. In yet another embodiment,“treat,”“treating,” or “treatment” refers to modulating a symptom of a disease or disorder of fat infiltration in muscle. In yet another embodiment,“treat,”“treating,” or“treatment” refers to preventing or delaying the onset or development or progression of a disease or disorder of fat infiltration in muscle.

Active Moieties: Compositions of Amino Acid Entities

US Patent Publication No. 2018/0207119, filed December 19, 2017, entitled AMINO ACID COMPOSITIONS AND METHODS FOR THE TREATMENT OF MUSCLE DISEASES AND DISORDERS, and US Patent Application Serial No. 15/847,289, filed December 19, 2017, entitled AMINO ACID COMPOSITIONS AND METHODS FOR THE TREATMENT OF LIVER DISEASES, each of which is specifically incorporated herein by reference in its entirety, disclose compositions of amino acid entities, i.e., Active Moieties, which may reduce fat infiltration in muscle, as can be shown by the methods herein described.

In some embodiments, the composition comprises a leucine amino acid entity, an arginine amino acid entity, a glutamine amino acid entity; and an antioxidant or reactive oxygen species (ROS) scavenger (e.g., a N-acetylcysteine (NAC) entity, e.g., NAC).

In some embodiments, the composition further comprises one or more essential amino acid (EAA)-entities. In some embodiments, the EAA-entities are chosen from one, two, three, or more (e.g., all) of a histidine (H)-amino acid-entity, a lysine (K)-amino acid-entity, a phenylalanine (F)-amino acid-entity, and a threonine (T)-amino acid-entity.

In some embodiments, the composition is capable of improving one or more

physiological symptoms selected from immobilization, malnutrition, fasting, aging, autophagy, reduced protein synthesis, anabolic resistance, neuromuscular junction integrity, insulin resistance, decreased mitochondrial biogenesis, anaplerosis, myogenesis, or an energy deficit.

As already noted, the present disclosure provides compositions that can include a leucine amino acid entity, an arginine amino acid entity, a glutamine amino acid entity; and an antioxidant or reactive oxygen species (ROS) scavenger, e.g., a N-acetylcysteine (NAC) entity, e.g., NAC. In some embodiments, the total weight of the leucine amino acid entity, arginine amino acid entity, glutamine amino acid entity; and ROS scavenger, e.g., a N- NAC entity, e.g., NAC, can be greater than the total wt. of other amino acid entities in the composition (e.g., Active Moiety).

In certain embodiments, two, three, or more (e.g., all) of methionine (M), tryptophan (W), valine (V), or cysteine (C) may be absent from the composition (e.g., Active Moiety), or if present, are present at less than 1 weight (wt.) %, less than 0.5 wt. %, or less than 0.1 wt. %, each of the foregoing on a dry weight basis. In some embodiments, methionine, tryptophan, valine, or cysteine, if present, may be present in an oligopeptide, polypeptide, or protein, with the proviso that the protein is not whey, casein, lactalbumin, or any other protein used as a nutritional supplement, medical food, or similar product, whether present as intact protein or protein hydrolysate.

In some embodiments, one or both of the arginine amino acid entity and the glutamine amino acid entity are present at a higher amount (wt. %) than the leucine amino acid entity. The arginine amino acid entity can be present, e.g., at an amount of at least 2 wt. %, at least 3 wt. %, at least 4 wt. %, at least 5 wt. %, at least 6 wt. %, at least 7 wt. %, or at least 8 wt. % greater than the leucine amino acid entity. The glutamine amino acid entity can be present, e.g., at an amount of at least 2 wt. %, at least 3 wt. %, at least 4 wt. %, or at least 5 wt. % greater than the L- leucine amino acid entity.

The weight ratio of a particular amino acid or particular amino acids in a composition or mixture of amino acids, i.e., in an Active Moiety, is the ratio of the weight of the particular amino acid or amino acids in the composition or mixture compared to the total weight of amino acids present in the composition or mixture. This value is calculated by dividing the weight of the particular amino acid or of the particular amino acids in the composition or mixture by the weight of all amino acids present in the composition or mixture and multiplying times 100. Percent weight on a dry weight basis refers to the percent weight of solid materials, which is relevant because as exemplified herein the Active Moiety composition may be dissolved or suspended in a liquid, particularly water, which provides considerable additional weight to a final liquid formulation.

The Active Moiety may further comprise additional branched-chain amino acid (BCAA)- entities, e.g., one or both of an isoleucine amino acid-entity and a valine amino acid-entity;

alternatively, both the isoleucine amino acid-entity and the valine amino acid-entity are present. The leucine amino acid entity can be present at a higher amount (% by weight) than one or both of the isoleucine amino acid-entity and the valine amino acid-entity (e.g., the leucine entity is present at an amount of at least 10 wt. %, at least 15 wt. %, at least 20 wt. %, at least 25 wt. %, at least 30 wt. %, at least 35 wt. %, at least 40 wt. %, at least 45 wt. %, or at least 50 wt. % greater than one or both of the isoleucine amino acid-entity and the valine amino acid-entity).

The composition can further comprise one or more essential amino acid (EAA)-entities.

In certain embodiments the EAA-entities are chosen from one, two, three, or four of a histidine amino acid-entity, a lysine amino acid-entity, a phenylalanine amino acid-entity, and a threonine amino acid-entity.

If present, the histidine amino acid-entity can be present in an amount of at least 0.5 wt. %, at least 0.6 wt. %, at least 0.7 wt. %, at least 0.8 wt. %, at least 0.9 wt. %, at least 1.0 wt. %, at least 1.1 wt. %, at least 1.2 wt. %, at least 1.3 wt. % or at least 1.4 wt. % of the composition on a dry weight basis.

If present, the lysine amino acid-entity can be present in amount of at least 2 wt. %, at least 3 wt. %, at least 4 wt. %, at least 5 wt. %, or at least 6 wt. % of the composition on a dry weight basis.

If present, the phenylalanine amino acid-entity can be present in an amount of at least 0.5 wt. %, at least 0.6 wt. %, at least 0.7 wt. %, at least 0.8 wt. %, at least 0.9 wt. %, at least 1.0 wt. %, at least 1.1 wt. %, at least 1.2 wt. %, at least 1.3 wt. % or at least 1.4 wt. % of the composition on a dry weight basis. If present, the threonine amino acid-entity can be present in amount of at least 0.5 wt. %, at least 1 wt. %, at least 1.5 wt. %, at least 2 wt. %, at least 2.5%, or at least 3 wt. % of the composition on a dry weight basis.

The histidine amino acid entity, lysine amino acid entity, phenylalanine amino acid entity, and threonine amino acid entity can all be present in the composition, including in the wt. % set forth in the preceeding four paragraphs.

The weight (wt.) ratio of the leucine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, and the NAC-amino acid entity in the Active Moiety can be about 1-3 : 2-4 : 2-4 : 0.1-2.5. In certain embodiments, the wt. ratio of the leucine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, and the NAC-amino acid entity is about 2 : 3 : 2.66 : 0.3. In certain embodiments, the wt. ratio of the leucine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, and the NAC-amino acid entity is about 2 : 3 : 2.66 : 0.6.

In some embodiments, the composition comprises a ratio of branched-chain amino acids to total amino acids of about 4:7 to about 1:2.

In some embodiments, the wt. ratio of the leucine amino acid entity, the isoleucine amino acid entity, the valine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, the NAC-amino acid entity, the histidine amino acid entity, the lysine amino acid entity, the phenylalanine amino acid entity, and the threonine amino acid entity is about 1-3 : 0.5-1.5 : 0.5- 1.5 : 2-4 : 2-4 : 0.1-0.5 : 0.1-0.5 : 0.2- 1.0 : 0.1-0.5 : 0.2-0.7. In certain embodiments, the wt. ratio of the leucine amino acid entity, the isoleucine amino acid entity, the valine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, the NAC-amino acid entity, the histidine amino acid entity, the lysine amino acid entity, the phenylalanine amino acid entity, and the threonine amino acid entity is about 2.0 : 1.0 : 1.0 : 3.0 : 2.66 : 0.3: 0.16 : 0.7 : 0.16 : 0.34. In certain embodiments, the wt. ratio of the leucine amino acid entity, the isoleucine amino acid entity, the valine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, the NAC-amino acid entity, the histidine amino acid entity, the lysine amino acid entity, the phenylalanine amino acid entity, and the threonine amino acid entity is about 2.0 : 1.0 : 1.0 : 3.0 : 2.66 : 0.3: 0.16 : 0.7 : 0.16 : 0.68.

In some embodiments, the total wt. of amino acids present in a unit dose of an Active Moiety is between about 4 g and about 80 g. In certain embodiments, the total wt. of amino acids present is about 6 g, about 18 g, about 24 g, about 48 g, about 68 g, or about 72 g in a unit dose of an active moiety. In an example, a unit dose of the Active Moiety comprises at least 1 g of the leucine amino acid entity, at least 0.5 g of the isoleucine amino acid entity, at least 0.5 g of the valine amino acid entity, at least 1.5 g of the arginine amino acid entity, at least 1.33 g of the glutamine amino acid entity, at least 0.15 g of the NAC-amino acid entity, at least 0.08 g of the histidine amino acid entity, at least 0.35 g of the lysine amino acid entity, at least 0.08 g of the phenylalanine amino acid entity, and at least 0.17 g of the threonine amino acid entity. In some embodiments, the composition comprises at least 1 g of the leucine amino acid entity, at least 0.5 g of the isoleucine amino acid entity, at least 0.5 g of the valine amino acid entity, at least 1.5 g of the arginine amino acid entity, at least 1.33 g of the glutamine amino acid entity, at least 0.3 g of the NAC-amino acid entity, at least 0.08 g of the histidine amino acid entity, at least 0.35 g of the lysine amino acid entity, at least 0.08 g of the phenylalanine amino acid entity, and at least 0.17 g of the threonine amino acid entity. Alternatively, the foregoing Active Moiety comprises 0.3 g of NAC instead of 0.15 g. Multiples of all these amounts in unit doses of the Active Moiety are also contemplated, e.g., 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, etc.

Preferably, at least one amino acid entity is a free amino acid, e.g., one or more (e.g., all) amino acid entities are a free amino acid. In some embodiments, the leucine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, and the NAC-amino acid entity is a free amino acid entity. In certain embodiment, the leucine amino acid entity, the isoleucine amino acid entity, the valine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, and the NAC-amino acid entity a free amino acid. In certain embodiments, the leucine amino acid entity, the isoleucine amino acid entity, the valine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, the NAC-amino acid entity, the histidine amino acid entity, the lysine amino acid entity, the phenylalanine amino acid entity, and the threonine amino acid entity is a free amino acid.

Alternatively, at least one amino acid entity is in a salt form, e.g., one or more (e.g., all) of the amino acid entities is in a salt form. In some embodiments, wherein the leucine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, and the NAC-amino acid entity is in a salt form. In certain embodiments, the leucine amino acid entity, the isoleucine amino acid entity, the valine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, and the NAC-amino acid entity is in a salt form. In certain embodiments, the leucine amino acid entity, the isoleucine amino acid entity, the valine amino acid entity, the arginine amino acid entity, the glutamine amino acid entity, the NAC-amino acid entity, the histidine amino acid entity, the lysine amino acid entity, the phenylalanine amino acid entity, and the threonine amino acid entity is in a salt form.

In some embodiments, the Active Moiety comprises a combination of 4 to 20 different amino acid entities, e.g., 5 to 15 different amino acid entities. In other embodiments, the Active Moiety consists of 4 to 16 different amino acid entities; more particularly, 5 to 15 different amino acid entities. For example, the Active Moiety can further comprise one or more (e.g., all) or more of serine, glycine, glutamine, HMB, citrulline, glutamine, L-cysteine, cystine, or glutathione.

In some embodiments, the composition comprises arginine, glutamine, N-acetylcysteine; a BCAA chosen from one, two, or all of leucine, isoleucine, and valine; and an essential amino acid EAA chosen from one, two, or all of histidine, lysine, phenylalanine, and threonine.

An aspect of the present disclosure provides a composition comprising an Active Moiety comprised of free amino acids and one or more pharmaceutically acceptable excipients, such that the amino acids include leucine, isoleucine, valine, arginine, glutamine, N-acetylcysteine, histidine, lysine, phenylalanine, and threonine.

An aspect of the present disclosure provides a composition comprising an Active Moiety consisting of free amino acids and one or more pharmaceutically acceptable excipients, such that the amino acids consist of leucine, isoleucine, valine, arginine, glutamine, N-acetylcysteine, histidine, lysine, phenylalanine, and threonine.

An exemplary Active Moiety can include leucine, isoleucine, valine, arginine HC1, glutamine, N-acetylcysteine, histidine, lysine, phenylalanine, and threonine as its defined amino acid components in a wt. ratio of 2.0 : 1.0 : 1.0 : 3.62 : 2.66 : 0.3: 0.16: 0.7: 0.16 : 0.34 (Table 3). The Active Moiety can include leucine, isoleucine, valine, arginine, glutamine, N-acetylcysteine, histidine, lysine, phenylalanine, and threonine as its defined amino acid components in a wt. ratio of 2.0 : 1.0 : 1.0 : 3.0 : 2.66 : 0.3: 0.16: 0.7: 0.16 : 0.34. Table 3. Exemplary amino acid components of the composition.

The composition can be administered in packets, e.g., packets containing about 6 g total amino acids as exemplified.

The composition can be administered three times daily at a dose of about 6 g of Active Moiety, i.e., total amino acids. In some embodiments, about 18 g, about 22, about 24 g, about 68 g or about 72 g total amino acids is administered per day. In an example, the composition is administered three times daily at a dose of about 24 g total amino acids for a total of about 72 g total amino acids is administered per day.

Methods of Treatment with an Active Moiety to Reduce Fat Infiltration In Muscle

The Active Moiety composition as described herein can be administered to reduce fat infiltration in muscle. Fat infiltration in muscle is an important predictor of muscle function and mobility, independent of as well as associated with other diseases and disorders of muscle atrophy (see, e.g,., Addison eta 1., 2014, Int. J. Endocrinology,

http://dx.doi .org/10.1 155/2014/309570) and has been identified as a complication of cirrhosis independently of other co-morbidities, e.g., sarcopenia (see, e.g., Bhanji et ah, 2018, Hep. Intk, Fat infiltration has an impact on muscle quality, so

identification of a therapeutic that can reduce fat infiltration in muscle is an important development. As shown in the Example, an exemplary Active Moiety, containing

LIVRQNacHFKT (using the single amino acid code with Nac meaning NAC as described above), showed significant reduction or even avoidance of fat infiltration in muscle that was immobilized for just seven days. This unprecedented observation for this class of active combinations of amino acid entities now identified as Active Moieties opens important new avenues of therapy with an Active Moiety for subjects in need of therapy, as well as methods of monitoring the effect of administration of an Active Moiety.

Thus, a method of the invention includes administration of an Active Moiety to treat subjects at risk of or who have fat infiltration in muscle, such as but not limited to subjects with chronic back pain (fat infiltration in paraspinal muscles); HIV patients (fat infiltration in locomotor muscles); spinal cord injury; stroke; COPD; hepatic encephalopathy; and muscle weakness associated with ageing.

In addition, lowering muscle fat is a means of improving glucose handling. Given a strong association between insulin resistance and muscle fat, lowering muscle fat improves glucose sensitivity. Glucose disposal is a function of muscle and so lowering fat infiltration in muscle improve a muscle function of sequestering glucose.

Increased infiltration by inter- and intramuscular fat (myosteatosis), in conjunction with reduced muscle mass (myopenia), is recognized as a poor prognostic indicator in patients with cancer (Malietzis et ah, Ann Surg. 2016 Feb;263(2):320-5). Significantly shorter cancer- specific survival and overall survival times were identified for the myosteatosis versus the

nonmyosteatosis group in a study of patients who underwent curative colorectal cancer surgery (Sueda et ah, Dis Colon Rectum. 2018 Mar;6l(3):364-374). Myosteatosis, characterized by inter- and intramyocellular fat deposition, is strongly related to poor overall survival after surgery for periampullary cancer (https://doi.Org/l0.l0l6/j.hpb.20l8.02.378). Thus, in another embodiment, reduction of fat infiltration in muscle by administration of an Active Moiety improves outcomes for treatment of cancer, such as surgery for colorectal cancer and for periampullary cancer.

Myosteatosis is independently associated with end-stage liver disease (ESLD), which is inclusive of, but not limited to, hepatic encephalopathy, variceal bleeding, portal hypertension, ascites, infection risk, sepsis, all-cause hospitalization, and all-cause and liver-related mortality. Subjects who have increased levels of fat infiltration in muscle may not have significant increase in BMI, sarcopenia, or other overt conditions. Accordingly, the invention provides a method of treating a patient who suffers from cirrhosis without sarcopenia by administering an effective amount of an Active Moiety that reduces fat infiltration in muscle. In another aspect, the invention provides for treating a patient with hepatic encephalopathy by administering an effective amount of an Active Moiety that reduces fat infiltration in muscle.

Patients with rotator cuff injury particularly benefit from the methods of the invention of administering an Active Moiety. Fat infiltration in the shoulder muscles is associated with poor outcome from rotator cuff injury (Melis et al., 2009, Orthopaedics & Tramatology: Surgery & Research 95:319-324). Rotator cuff injury includes tendon lesion of the supraspinatus, infraspinatus, and subscapularis. Fat infiltration can occur in any of the affected muscles. Fat infiltration above stage 2 (intermediate) according to the Goutallier classification (see Melis et al.) carries the risk of irreversible functional loss, and a key objective of an intervention during intermediate stages of muscle fatty infiltration is to prevent such permanent loss. Thus, the present invention provides for treating subjects suffering from rotator cuff injury with an Active Moiety to prevent or delay onset of Goutallier Stage 2 fat infiltration, especially prior to surgery to repair the injury. Alternatively, the present invention provides for treating subjects suffering from rotator cuff injury who have Goutallier Stage 2 or greater fat infiltration to reduce the degree of fat infiltration, and thus the Goutallier Stage, prior to surgery. In yet another alternative, the invention provides for treating a subject suffering from a rotator cuff injury with an Active Moiety in addition to medical treatment; medical treatment of rotator cuff injury includes rest, adaptation of daily and occupational movements, rehabilitation, NSAIDS, antalgics, physical therapy, infiltrations, etc. Usually surgery for a rotator cuff injury is surgery to repair a torn tendon, and can be arthroscopic surgery or normal surgery.

The invention is especially useful for treating elderly subjects with a rotator cuff injury, who are at greater risk for more faster fat infiltration and progression to the intermediate Goutallier Stage 2 and beyond to more severe fat infiltration (Stages 3 and 4). Elderly subjects 50 years old or older benefit; subjects 55 years old or older can have even greater benefit; and subjects 60 years old or older even greater benefit than those who are 55 or older, since at each advanced age group susceptibility to fat infusion increases. Each of the foregoing treatments of rotator cuff injury may include determining the degree of fat infiltration in muscle, e.g., to determine the Goutallier stage; to show changes in Goutallier stage with therapy by administering an Active Moiety of the invention; to obtain a prognosis of therapy, such as surgery; to establish an appropriate time for surgery; or to determine that surgery is unnecessary.

Treatment with an Active Moiety accompanied by evaluating fat infiltration in muscle

In other conditions, administering the Active Moiety to improve, e.g., enhance, muscle function, e.g., in a patient with a muscle disease or disorder, includes evaluating fat infiltration in muscle. The present disclosure also provides a method for treating one or more (e.g., all) physiological symptoms selected from immobilization, malnutrition, fasting, aging, autophagy, reduced protein synthesis, anabolic resistance, neuromuscular junction integrity, insulin resistance, decreased mitochondrial biogenesis, anaplerosis, or an energy deficit, in each case along with evaluating fat infiltration in muscle. The method includes administering to a subject in need thereof an effective amount of a composition as set forth hereinabove. Therapeutic treatment according to the invention can be achieved in a subject who has a muscle disease, for example, muscle atrophy, sarcopenia, muscle deterioration, muscle decay, cachexia, drug- induced myopathy, muscular dystrophy, or myopenia. The muscle disease or disorder can be a dystrophy, such as a myotonic dystrophy. For example, the muscle disease or disorder can be DM1.

Alternatively, the muscle disease or disorder can be a drug-induced myopathy, e.g., a statin-induced myopathy; a steroid-induced myopathy; an immunosuppressant-induced myopathy; a chemotherapeutic-induced myopathy; or an alcohol-induced myopathy. In each case, administration of an Active Moiety is accompanied by evaluating fat infiltration in muscle.

In addition, the subject can have a fracture or other trauma other than rotator cuff injury.

In some embodiments, the method includes administering to a subject in need thereof an effective amount of the composition to treat a food deficiency, e.g., malnutrition or fasting; aging; autophagy; reduced protein synthesis; anabolic resistance; junction integrity (e.g., neuromuscular junction integrity); decreased mitochondrial biogenesis; anaplerosis. In each such foregoing case, treatment is accompanied by evaluating fat infiltration in muscle. In some embodiments, the subject has not received prior treatment with an Active Moiety (e.g., a naive subject) accompanied by evaluating fat infiltration in muscle.

In some embodiments, the subject has muscle weakness, e.g., muscle weakness of one, two, or more (e.g., all ) of skeletal muscle, cardiac muscle, or smooth muscle. In certain embodiments, the subject has muscle weakness in one, two, three, four, five, six, or more (e.g., all) of a neck muscle, a torso muscle, an arm muscle, a shoulder muscle, a hand muscle, a leg muscle, or a foot muscle. Fat infiltration in muscle can be evaluated daily, every 2-3 days, weekly, every two weeks, every three weeks, and every four weeks after treatment to determine or evaluate the degree of fat infiltration in muscle, particularly to determine that the degree of fat infiltration in muscle is reduced by treatment with an Active Moiety. In the case where administration of an Active Moiety precedes an elective procedure, such as orthopedic surgery, evaluating fat infiltration in muscle can be undertaken to determine that the fat fraction in muscle is unchanged from before the surgery, or even improved.

A subject who has had orthopedic surgery, e.g., knee surgery, or hip surgery, elbow surgery, or has worn a cast benefits from administration of an Active Moiety accompanied by evaluating fat infiltration in muscle. When surgery is elective, e.g., for knee or hip replacement (also called total knee arthroplasty and total hip arthroplasty, respectively) administration of the Active Moiety, evaluation of fat infiltration in muscle, or both can be done before surgery, e.g., one week before surgery or two weeks before surgery, or can be done after surgery, and preferably is done before and after surgery. In particularly, evaluation of fat infiltration in muscle, specifically muscle most impacted by the surgery or treated by the surgery, can be done daily, every 2-3 days, weekly, every two weeks, every three weeks, and every four weeks after surgery to determine or evaluate the degree of fat infiltration in muscle, particularly to determine that the degree of fat infiltration in muscle is reduced by treatment with an Active Moiety, and more particularly to determine that the fat fraction in muscle is unchanged from before the surgery, or even improved.

In some embodiments, the subject has a neuromuscular disorder, e.g., myasthenia gravis or Lambert-Eaton myasthenic syndrome.

In some embodiments, the subject has muscular dystrophy, e.g., Duchenne muscular dystrophy, Becker muscular dystrophy, facioscapulohumeral muscular dystrophy, or myotonic dystrophy. In some embodiments, the subject has an inflammatory myopathy, e.g., polymyositis or dermatomyositis.

In some embodiments, the subject has one, two, or more (e.g., all) of low sodium levels (e.g., hyponatremia), low potassium levels (e.g., hypokalemia), or a calcium deficiency or relatively high calcium levels (e.g., hypercalcemia).

In some embodiments, the subject has muscle weakness associated with nerve damage, e.g., neuralgia or peripheral neuropathy. In some embodiments, the subject has a bone weakness disease, e.g., osteomalacia, osteogenesis imperfecta, rickets, or Hypophosphatasia.

In some embodiments, the subject has experienced a stroke or a transient ischemic attack. In some embodiments, the subject has an autoimmune disease, e.g., Graves’ disease.

In some embodiments, the subject has hypothyroidism. In some embodiments, the subject has amyotrophic lateral sclerosis (ALS).

In some embodiments, administering the composition results in activation of muscle protein synthesis in the subject. In some embodiments, the composition also reduces muscle protein wasting.

In some embodiments, the composition results in an improvement in the degree of fat infiltration in muscle associated with one or both of immobilization or muscle disuse following injury in a subject. In some embodiments, the subject has had a surgery, e.g., rotator cuff surgery, knee surgery, or hip surgery, or has worn a cast, prior to administration of the composition. In some embodiments, the subject has had a hip fracture-related myopenia. In some embodiments, the subject has had a joint replacement. In some embodiments, the subject has had an injury repair surgery.

In some embodiments, the subject has ventilator-induced diaphragmatic dystrophy or ventilator-induced diaphragmatic dysfunction. In some embodiments, the subject has had one or both of ICU-acquired or bums-related myopathies.

In some embodiments, the subject has disease-related cachexia, e.g., a disease-related cachexia selected from chronic obstructive pulmonary disease (COPD), congestive heart failure (CHF), chronic kidney disease (CKD), and cancer.

In some embodiments, method of the invention further includes administration of a second agent. Such a second agent may exclude proteins, whether intact or in hydrolyzed form, such as whey, casein, lactalbumin, etc. The present disclosure also provides a method for reducing muscle atrophy comprising administering to a subject in need thereof an effective amount of a composition described herein. In each such case effectiveness of therapy includes reduction of fat infiltration in muscle, and may include determining the degree of fat infiltration in muscle.

The present disclosure also provides a composition described herein for use as a medicament for reducing fat infiltration in muscle.

The present disclosure provides a composition described herein for use as a medicament for reducing fat infiltration in muscle, which may be in conjunction with treating one or more symptoms selected from the group consisting of immobilization, injury, surgery, malnutrition, fasting, aging, autophagy, reduced protein synthesis, anabolic resistance, neuromuscular junction integrity, insulin resistance, decreased mitochondrial biogenesis, and anaplerosis.

Dosage Regimens

The composition can be administered to a human subject according to a dosage regimen described herein.

Doses can be administered, e.g., twice daily, three times daily, four times daily, five times daily, six times daily, seven times daily, or more. The composition can be administered for at least 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or 2 weeks. Depending on the condition being treated, the composition can be administered for at least 10 weeks, 11 weeks, 12 weeks, 13 weeks, 14 weeks, 15 weeks, 16 weeks, 17 weeks, 18 weeks, 19 weeks, 20 weeks, or longer. In some embodiments, the composition can be administered chronically, e.g., more than 30 days, e.g., 31 days, 40 days, 50 days, 60 days, 3 months, 6 months, 9 months, one year, two years, or three years). If the condition of fat infiltration in muscle is chronic and unremitting, the invention contemplates administering an Active Moiety indefinitely, e.g., for the life of the subject.

The Active Moiety can be administered at a dose of about 4 g and about 80 g total amino acids, e.g., once per day, twice per day, three times per day, four times per day, five times per day, or six times per day (e.g., three times per day). In some embodiments, the composition is administered at a dose of about 5 g to about 15 g, about 10 g to about 20 g, about 20 g to about 40 g, or about 30 g to about 50 g total amino acids, e.g., once per day, twice per day, three times per day, four times per day, five times per day, or six times per day (e.g., three times per day). The composition can be administered every 2 hours, every 3 hours, every 4 hours, every 5 hours, every 6 hours, every 7 hours, every 8 hours, every 9 hours, or every 10 hours to enhance muscle function in a subject (e.g., the subject has or is identified as having decreased muscle function due to aging, injury, atrophy, infection, or disease).

In some embodiments, the composition is administered prior to a meal (e.g., one, two, or more (e.g., all) of breakfast, lunch, or dinner). In some embodiments, the composition is administered conccurrent with a meal (e.g., one, two, or more (e.g., all) of breakfast, lunch, or dinner). In some embodiments, the composition is administered following a meal (e.g., one, two, or more (e.g., all) of breakfast, lunch, or dinner).

Production of Active Moiety and Pharmaceutical Compositions

Amino acids used to make the compositions may be agglomerated, and/or instantized to aid in dispersal and/or solubilization.

The amino acid compositions of the present disclosure may be made using amino acids and amino acid derivatives from the following sources, or other sources may used: e.g., FUSI- BCAA™ Instantized Blend (L- Leucine, L-Isoleucine and L-Valine in 2:1:1 weight ratio), FUSIL™ Instantized L-Leucine, L- Arginine HC1, L-Glutamine and other amino acids may be obtained from Ajinomoto Co., Inc; N-acetylcysteine may be obtained from Spectrum Chemical.

To produce the amino acid compositions of the instant disclosure, the following general steps may be used: the starting materials (individual amino acids and excipients) may be blended in a blending unit, followed by verification of blend uniformity and amino acid content, and filling of the blended powder into stick packs or other unit dosage form. The content of stick packs or other unit dosage forms may be dispersed in water at time of use for oral administration.

Pharmaceutical compositions of the present disclosure may be in a form suitable for oral use (for example aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for parental administration (for example as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular dosing or as a suppository for rectal dosing) or for enteral administration (for example via tube feeding). Generally, however, pharmaceutical compositions of the invention will be for oral administration.

Food supplement and medical nutrition compositions of the invention will be in a form suitable for oral administration. When combining raw materials, e.g., pharmaceutical grade amino acid entities and/or excipients, into a composition, contaminants may be present in the composition. A composition meets a standard for level of contamination when the composition does not substantially comprise (e.g., comprises less than 10, 5, 1, 0.1, 0.01, or 0.001% (w/w on a dry weight basis)) a contaminant. In some embodiments, a composition described in a method herein does not comprise a contaminant. Contaminants include any substance that is not deliberately present in the composition (for example, pharmaceutical grade amino acid entities and excipients, e.g., oral administration components, may be deliberately present) or any substance that has a negative effect on a product quality parameter of the composition (e.g., side effects in a subject, decreased potency, decreased stability/shelf life, discoloration, odor, bad taste, bad texture/mouthfeel, or increased segregation of components of the composition). In some embodiments, contaminants include microbes, endotoxins, metals, or a combination thereof. In some embodiments, the level of contamination, e.g., by metals, lecithin, choline, endotoxin, microbes, or other contaminants (e.g., contaminants from raw materials) of each portion of a composition is below the level permitted in food.

Excipients

The amino acid compositions of the present disclosure may be compounded or formulated with one or more excipients. Non-limiting examples of suitable excipients include a tastant, a flavorant, a buffering agent, a preservative, a stabilizer, a binder, a compaction agent, a lubricant, a dispersion enhancer, a disintegration agent, a flavoring agent, a sweetener, and a coloring agent.

In some embodiments, the excipient comprises a buffering agent. Non-limiting examples of suitable buffering agents include citric acid, sodium citrate, magnesium carbonate, magnesium bicarbonate, calcium carbonate, and calcium bicarbonate.

In some embodiments, the excipient comprises a preservative. Non-limiting examples of suitable preservatives include antioxidants, such as alpha-tocopherol and ascorbate, and antimicrobials, such as parabens, chlorobutanol, and phenol.

In some embodiments, the composition comprises a binder as an excipient. Non-limiting examples of suitable binders include starches, pregelatinized starches, gelatin,

polyvinylpyrolidone, cellulose, methylcellulose, sodium carboxymethylcellulose, ethylcellulose, polyacrylamides, polyvinyloxoazolidone, polyvinylalcohols, C 12-08 fatty acid alcohol, polyethylene glycol, polyols, saccharides, oligosaccharides, and combinations thereof.

In some embodiments, the composition comprises a lubricant as an excipient. Non limiting examples of suitable lubricants include magnesium stearate, calcium stearate, zinc stearate, hydrogenated vegetable oils, sterotex, polyoxyethylene monostearate, talc,

polyethyleneglycol, sodium benzoate, sodium lauryl sulfate, magnesium lauryl sulfate, and light mineral oil.

In some embodiments, the composition comprises a dispersion enhancer as an excipient. Non-limiting examples of suitable dispersants include starch, alginic acid, polyvinylpyrrolidones, guar gum, kaolin, xanthan gum, bentonite, purified wood cellulose, sodium starch glycolate, isoamorphous silicate, and microcrystalline cellulose as high HLB emulsifier surfactants.

In some embodiments, the composition comprises a disintegrant as an excipient. In some embodiments, the disintegrant is a non-effervescent disintegrant. Non-limiting examples of suitable non-effervescent disintegrants include starches such as com starch, potato starch, pregelatinized and modified starches thereof, sweeteners, clays, such as bentonite, micro crystalline cellulose, alginates, sodium starch glycolate, gums such as agar, guar, locust bean, karaya, pecitin, and tragacanth. In some embodiments, the disintegrant is an effervescent disintegrant. Non-limiting examples of suitable effervescent disintegrants include sodium bicarbonate in combination with citric acid, and sodium bicarbonate in combination with tartaric acid.

In some embodiments, the excipient comprises a flavoring agent. Flavoring agents can be chosen from synthetic flavor oils and flavoring aromatics; natural oils; extracts from plants, leaves, flowers, and fruits; and combinations thereof. In some embodiments, the flavoring agent is selected from cinnamon oils; oil of wintergreen; peppermint oils; clover oil; hay oil; anise oil; eucalyptus; vanilla; citrus oil such as lemon oil, orange oil, grape and grapefruit oil; and fruit essences including apple, peach, pear, strawberry, raspberry, cherry, plum, pineapple, and apricot.

In some embodiments, the excipient comprises a sweetener. Non-limiting examples of suitable sweeteners include glucose (corn syrup), dextrose, invert sugar, fructose, and mixtures thereof (when not used as a carrier); saccharin and its various salts such as the sodium salt;

dipeptide sweeteners such as aspartame; dihydrochalcone compounds, glycyrrhizin; Stevia Rebaudiana (Stevioside); chloro derivatives of sucrose such as sucralose; and sugar alcohols such as sorbitol, mannitol, sylitol, and the like. Also contemplated are hydrogenated starch hydrolysates and the synthetic sweetener 3,6-dihydro-6-methyl-l,2,3-oxathiazin-4-one-2,2- dioxide, particularly the potassium salt (acesulfame-K), and sodium and calcium salts thereof.

In some embodiments, the composition comprises a coloring agent. Non-limiting examples of suitable color agents include food, drug and cosmetic colors (FD&C), drug and cosmetic colors (D&C), and external drug and cosmetic colors (Ext. D&C). The coloring agents can be used as dyes or their corresponding lakes.

Particular excipients may include one or more of: citric acid, lecithin, (e.g. Alcolec F100), sweeteners (e.g. sucralose, sucralose micronized NF, acesulfame potassium (e.g. Ace-K)), a dispersion enhancer (e.g. xanthan gum (e.g. Ticaxan Rapid-3)), flavorings (e.g. vanilla custard #4306, Nat Orange WONF #1326, lime 865.0032U, and lemon 862.2169U), a bitterness masking agent (e.g. 936.2160U), and natural or artificial colorings (e.g. FD&C Yellow 6). Table 5 in the Example illustrates a formulation with such excipients.

Dietary Compositions

The Active Moiety including amino acid entities can be formulated and used as a dietary composition, e.g., chosen from a medical food, a functional food, or a supplement. In such an embodiment, the raw materials and final product should meet the standards of a food product. Such uses include improving health of a subject having or identified as suffering from infiltration of fat in muscle due to aging, injury, atrophy, infection, or disease. In some embodiments, the subject has or is identified as having muscle deterioration, muscle decay, muscle atrophy, cachexia, sarcopenia, steroid myopathy, or muscular dystrophy. In some embodiments, the subject has one or both of type 2 diabetes or a relatively high BMI.

In some embodiments, administration of the dietary composition results in an

improvement in one or more metabolic symptoms in the subject, e.g., one or more metabolic symptoms is selected from the following: increased free fatty acid and lipid metabolism, improved mitochondrial function, white adipose tissue (WAT) browning, decreased reactive oxygen species (ROS), increased levels of glutathione (GSH), decreased hepatic inflammation, decreased hepatocyte ballooning, improved gut barrier function, increased insulin secretion, or glucose tolerance. In certain embodiments, administration of the composition results in an improvement in one or more metabolic symptoms after a treatment period of 24 hours.

Biomarkers

Any of the methods disclosed herein can include evaluating or monitoring the

effectiveness of administering a composition described herein to a subject by determining the degree of infiltration of fat in muscle in the subject, e.g., with CT or MRI. The subject may be in need of muscle function enhancement (e.g., a subject having muscle deterioration, muscle decay, muscle atrophy, cachexia, sarcopenia, drug-induced myopathy, muscular dystrophy, or myopenia). The effectiveness to the composition in treating a subject can further comprise a measure of the levels of one or more (e.g., all) of the following:

a) myostatin;

b) myoglobin;

c) Cortisol- AM;

d) C-reactive protein;

e) insulin;

f) cytokines (e.g., one or more (e.g., all) of IL-1A RBM, IL-1RA, IL-l RI, IL-l RII, IL-

12, IL-18, or MCP-l);

g) GDF-l l;

h) P3NP;

i) IGF-l;

j) IGFBP1;

k) IGFBP3;

l) FGF21;

m) DHEAS;

n) mTORCl ;

o) Gcn2; or

p) AMP-activated protein kinase (AMPK).

In some embodiments of any of the methods disclosed herein, the measure of one or more of a)-p) is obtained from a sample acquired from the subject. In some embodiments, the subject is evaluated prior to receiving, during, or after receiving, the composition.

In some embodiments, administration of the composition to the subject results in a decrease in levels of one or more (e.g., all) of myoglobin, myostatin, GDF-l l, cortisol-AM, C- reactive protein, insulin, or cytokines (e.g., one or more (e.g., all) of IL-1A RBM, IL-1RA, IL-l RI, IL-l RII, IL-12, IL-18, or MCP-l) in the subject (Table 4). In some embodiments, administration of the composition to the subject results in an increase in levels of one or more (e.g., all) of P3NP, IGF-l, IGFBP1, IGFBP3, FGF-21, DHEAS, or mTORCl in the subject (Table 4).

Table 4. Additional biomarkers to determine effect of the composition on muscle biology.

EXAMPLE

The Example below is set forth to aid in the understanding of the invention, but is not intended to, and should not be construed to, limit its scope in any way.

Example 1. Treatment of Immobilization in Subjects with an Amino Acid Composition

Not Only Reduces Loss of Muscle Mass and Function, But Surprisingly

Reduces Fat Infiltration.

The study described herein features the administration of a composition including amino acids to healthy subjects undergoing unilateral knee immobilization. The goal of this study was to determine the impact of an amino acid composition on muscle atrophy after 7 days of single leg immobilization and 14 days of recovery post-immobilization. The composition included about 1 g of L-leucine, about 0.5 g of L-isoleucine, about 0.5 g of L-valine, about 1.5 g of L- arginine (or 1.81 g of L-arginine HC1), about 1.33 g of L-glutamine, about 0.15 g of N- acetylcysteine, about 0.08 g of L-histidine, about 0.35 g of L-lysine, about 0.08 g of L- phenylalanine, and about 0.17 g of L-threonine per stick packet for administration in four stick packs three times per day (e.g., a total of about 68 or 72 g per day, or about 23 g or 24 g three times per day). The composition also included excipients as shown in Table 5.

Table 5. Ingredient contents in each stick pack.

In the study, subjects received the amino acid composition three times daily for 28 days. Amino acids were provided in powder form to be dissolved in 8 oz. of water. Participants underwent single-leg immobilization for 7 days (days 8-15) during the 28-day study period. An immobilization device was used for 7 days of single-leg immobilization of the dominant knee (based on maximal isometric leg strength) with a knee brace worn in a fixed flexion position at 140° (e.g., a Breg brace).

Control subjects received placebo three times daily for 28 days. Placebo consisted of an amount of maltodextrin (NF grade) equivalent in caloric content to the amount of amino acids administered, with the same excipients, dissolved in 8 oz. of water.

The primary outcome measure of this study was safety and tolerability. In addition, muscle disuse atrophy, in particular, the impact of the amino acid formulation on muscle atrophy after 7 days of single leg immobilization was studied. The secondary outcome measures included muscle function based on knee strength, muscle cross-section area and volume, muscle fiber quality, and lean muscle mass. The percentage change in lean muscle mass in the subjects was determined using dual-energy x-ray absorptiometry (DEXA). The percentage change in maximum torque as measured using a BioDex machine (measured in Newton-meters) and percentage change in the time to maximum torque (measured in seconds) were also assessed. Muscle biopsies were performed to determine muscle fiber cross-sectional area (CSA). Muscle size was assessed via MRI. Muscle health was assessed by electrical impedance myography (EIM) measurements. Assessments were performed at baseline (day 1), pre-immobilization (day 8), post-immobilization (day 15), and recovery (day 28).

More specifically, MRIs were performed at Days 1, 8, 15, and 28. Axial (transverse) images were obtained from both thighs from the distal end of the femur to the greater trochanter using GE high fidelity 3T magnet. A fast-recovery, fast spin echo pulse sequence was used, along with IDEAL (iterative decomposition of water and fat with echo asymmetry and least- squares estimation) post-processing to obtain water-only, fat-only, in-phase and out-of-phase images of the thighs. The following parameters were used: TR = 2000 msec, TE = 30 msec, refocusing flip angle = 111 degrees, echo train length = 6, ASSET (parallel imaging factor) = 2, field of view = 42 x 21 cm, acquisition matrix = 512 x 256, 3-mm slice thickness, 0-mm slice gap. A total of approximately 160 slices were acquired, but varied depending on length of the thigh. The acquisition was done in two sections, a lower stage, and an upper stage. Total scan time for both stages was approximately 11 minutes. The scans were uploaded onto Analyze Pro software. The 50% region between the greater trochanter of the hip and lateral epicondyle of the knee were used for analysis. The segmentation features of the software were used to differentiate between the bone, fat, right muscle, right quadriceps, left muscle and left quadriceps. Then every third slice in the 50% region was manually traced for the quadriceps muscles of both legs. The highest number from these measurements was taken as the peak quadriceps cross-sectional area. The software was then able to take every third slice that was manually measured and extrapolate that data for every slice in the 50% region to get an estimate of quadriceps volume. CSA was expressed in mm2 and muscle volume in mm3. Protocol adapted from Reeder et ah, 2005.

To obtain independent verification of the imaging data, DIXON sequences of the upper and lower thighs were securely transferred to the Image Analysis Group (IAG, London, UK) for whole muscle volume analysis and an additional analysis to measure intramuscular fat fraction. Given the water and fat images it is possible to generate a Fat Fraction (FF) image as:

FF = F/(W+F), where F = fat, and W = water.

IAG calculated these images and added them to the individual DICOM studies. As the base images can give spurious regions of high fat fraction due to noise, a thresholding filter was used to reduce these small peripheral artefacts and minimize noise in regions where both the fat and water signals are small. Segmentations were carried out on the upper thigh FF images. The segmentation was from the middle of the thigh towards the pelvis for 20 slices. The segmentation was carried out manually from each sequence. Once segmented, the slice ROIs were grouped to form a volume ROI and the statistics automatically calculated.

Key criteria for selecting subjects included the following: 1) generally healthy, non smoking; 2) willing and able to provide informed consent; 3) men age 20-45 years; and 4) BMI between 25 and 35 kg/m 2 . Exclusion Criteria included the following: 1) smokers; 2) subject has any concurrent medical, orthopedic, or psychiatric condition that, in the opinion of the investigator, would compromise his/her ability to comply with the study requirements; 3) history of cancer within the last 5 years, except basal cell carcinoma, non-squamous skin carcinoma, prostate cancer, or carcinoma in situ with no significant progression over the past 2 years; 4) significant orthopedic, cardiovascular, pulmonary, renal, liver, infectious disease, immune disorder (requiring ongoing medical care), or metabolic/endocrine disorder (e.g., diabetes, high cholesterol, elevated fasting blood sugar) or other disease that would preclude oral protein supplement ingestion and/or assessment of safety and study objectives; 5) any cachexia-related condition (e.g., relating to cancer, tuberculosis, or human immunodeficiency virus infection and acquired immune deficiency syndrome) or any genetic muscle diseases or disorders; 6) current illnesses that could interfere with the study (e.g. prolonged severe diarrhea, regurgitation, or difficulty swallowing); 7) subject participated in a study of an investigational product less than 60 days or 5 half-lives of the investigational product, whichever is longer, before enrollment in this study; 8) hypersensitivity to any of the components of the test product; 9) excessive alcohol consumption (>21 units/week); 10) known sensitivity or allergy to amino acids or any ingredient in the test formulations; 11) prior gastrointestinal bypass surgery (e.g., lapband surgery), irritable bowel disease, or irritable bowel syndrome; 12) history of bleeding diathesis, platelet or coagulation disorders, or antiplatelet/anticoagulation therapy (up to 81 mg of baby aspirin per day taken as a prophylactic is permitted); 13) personal or family history of clotting disorder or deep vein thrombosis; 14) concomitant use of corticosteroids, testosterone replacement therapy (ingestion, injection, or transdermal), any anabolic steroid, creatine, whey protein supplements, casein, or branched-chain amino acids (BCAAs) within 45 days prior to screening; 15) contraindications to an MRI scan (e.g. subjects with non-removable ferromagnetic implants, pacemakers, aneurysm clips or other foreign bodies, or subjects with claustrophobic symptoms that would contraindicate an MRI scan); 16) hemoglobin less than l l.5mg/dl at screening; or 17) platelets less than l50,000/uL (l50xl09/L) at screening.

The findings from this study suggest that the decline in lean leg mass as a result of unilateral limb immobilization (i.e. disuse atrophy), including reduction in fat infiltration into the muscle, was attenuated in those that received the LIVRQNACHKFT amino acid combination, as compared to those that received placebo . These results in subjects undergoing a unilateral limb immobilization suggest that the amino acid combination attenuated this decline in lean mass of the immobilized leg, while preserving muscle strength. The immobilized leg in the placebo administered groups did not recover their lean mass to the post-immobilized or the pre immobilized state during the two week recovery period. By contrast, administration of the amino acid combination maintained and/or improved the lean leg mass within this two week recovery period to that of the post and pre-immobilization. The decline in muscle strength seen after a week of unilateral limb immobilization in the placebo group was also attenuated by the amino acid combination. The non-immobilized leg in either the Placebo or the

LIVRQNACHKFT amino acid administered group did not appear to lose their lean leg mass nor their muscle strength to the same extent as the corresponding immobilized leg during the knee brace period, as expected of an appropriate control.

CSA of specific fibers within the vastus lateralis was preserved during immobilization with LIVRQNACHKFT vs. Pbo administration. One week of immobilization led to a 2.2% (± 4.0) decrease in fiber CSA (Figure 1). Consistent with the existing literature, in the Pbo group, muscle disuse tended to result in a preferential loss of Type II vs. Type I fibers (4.5 ± 4.8% loss for Type II vs. no change for Type I). By contrast, LIVRQNACHKFT administration preserved the cross- sectional area of both fiber types during immobilization. Quantification of CSA is shown in Figure 1 , with the following changes observed between LIVRQNACHKFT and Pbo: increase of 4.l-fold for total fibers; 1.7-fold for Type II fibers; l2-fold for Type I fibers (P = 0.08). Of note,

LIVRQNACHKFT had a particularly pronounced effect on the slow twitch type I fibers, in not only preserving them, but tended to induce their growth (13.3 ± 7.4% increase in Type I fiber CSA from Day 8 to Day 15).

The unprecedented observation of LIVRQNACHKFT’s effect on the oxidative, slow twitch Type I fibers could suggest an impact on insulin sensitization, and the latter has been closely associated with muscle fat infiltration (Albu et al. 2005). Consistent with this unprecented effect on Type I muscle fibers, LIVRQNACHKFT administration significantly attenuated muscle fat infiltration during limb immobilization (Figures 2A and 2B). Representative images depicting fat fraction (FF) changes show that in a subject administered Pbo (Figure 2A), the non-immobilized leg had no change in FF, while the immobilized leg had increased FF and decreased muscle mass between Day 8 and 15. By contrast, in a subject administered LIVRQNACHKFT (Figure 2A), the immobilized leg had lower fat fraction, and a higher muscle content following immobilization, illustrating LIVRQNACHKFT anti-atrophic effects. These FF changes were quantified across all subjects and results are plotted in Figure 2B. Percent change in quadriceps muscle fat fraction on Day 15 vs. Day 8 was +12.8 + 6.1% in Pbo vs. -0.41 + 3.07% in LIVRQNACHKFT (P = 0.018) in the immobilized leg. As expected, the non-immobilized leg had significantly less to no muscle fat infiltration: 1.76+2.6% (Pbo) vs. -2.05+2.4% (LIVRQNACHKFT), with no statistical difference between the groups (P = 0.479, Figure 2B).

Consistent with the literature (Tarulli et al. 2009), we observed decreases in phase, maximum reactance, and reactance slope (these parameters are considered to be reflective of muscle health, Rutkove 2009) during limb immobilization (i.e. from Day 8 to 15) in the Pbo group (Figure 3).

By contrast, LIVRQNACHKFT administration resulted in the attenuation, if not a numerical increase in these parameters during immobilization (with relative differences versus Pbo ranging from 115% to 155%). During the recovery phase, these parameters returned to pre-immobilization levels in both groups (Day 28 vs. Day 8), but these parameters tended to be higher in the

LIVRQNACHKFT group compared to Pbo, with relative differences of 56%, 60%, and 70% for phase, max reactance, and reactance slope, respectively.

While the invention has been particularly shown and described with reference to a preferred embodiment and various alternate embodiments, it will be understood by persons skilled in the relevant art that various changes in form and details can be made therein without departing from the spirit and scope of the invention.

All references, issued patents and patent applications cited within the body of the instant specification are hereby incorporated by reference in their entirety, for all purposes.