Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
LIVER DISEASE
Document Type and Number:
WIPO Patent Application WO/2020/018974
Kind Code:
A1
Abstract:
Methods of treating a liver disease or condition, or a symptom thereof, in a subject in need of treatment, are disclosed and described. One method comprises orally administering to a subject, a pharmaceutical composition having an amount of a testosterone, or an ester thereof, sufficient to treat the liver disease or condition, or symptom thereof.

Inventors:
PATEL MAHESH V (US)
CHIDAMBARAM NACHIAPPAN (US)
NACHAEGARI SATISH K (US)
BYRNE BURKE (US)
KIM KILYOUNG (US)
BAKER JONATHAN A (US)
Application Number:
PCT/US2019/042709
Publication Date:
January 23, 2020
Filing Date:
July 19, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
LIPOCINE INC (US)
International Classes:
G01N33/74; H01J49/00
Domestic Patent References:
WO2014049131A12014-04-03
Foreign References:
US20160361322A12016-12-15
US20160184320A12016-06-30
Other References:
NEFF ET AL.: "Topical testosterone treatment for chronic allograft failure in liver transplant recipients with recurrent hepatitis C virus", TRANSPLANT PROC., vol. 36, no. 10, December 2004 (2004-12-01), pages 3071 - 4, XP004730815, DOI: 10.1016/j.transproceed.2004.11.004
SINCLAIR ET AL.: "Testosterone in men with advanced liver disease: Abnormalities and implications", JOURNAL OF GASTROENTEROLOGY AND HEPATOLOGY, vol. 30, 2015, XP055676755
YURC I ET AL.: "Effects of testosterone gel treatment in hypogonadal men with liver cirrhosis", CLIN RES HEPATOL GASTROENTEROL. 2011 DEC, vol. 35, no. 12, 9 November 2011 (2011-11-09), pages 845 - 54, XP055676760, DOI: 10.1016/j.clinre.2011.09.005
"Testosterone treatment of men with alcoholic cirrhosis: a double-blind study", HEPATOLOGY, vol. 6, no. 5, September 1986 (1986-09-01), pages 807 - 13, XP055676765
CHAUDHURY ET AL.: "Testosterone in Men With Chronic Hepatitis C Infection and After Hepatitis C Viral Clearance", TESTOSTERONE AND HEPATITIS C INFECTION, pages 571 - 576, XP055676781, DOI: 10.1093/cid/ciy965
See also references of EP 3824296A4
Attorney, Agent or Firm:
OSBORNE, David W. (US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAIMED IS:

1. A method of treating a liver disease or condition, or a symptom thereof, in a subject in need of treatment, said method comprising orally administering to a subject a pharmaceutical composition having an amount of a testosterone, or an ester thereof, sufficient to treat the liver disease or condition, or symptom thereof.

2. The method of claim 1 wherein said method ameliorates the level of one or more biomarkers in said subject compared to the baseline value.

3. The method of claim 1 wherein the subject is a male.

4. The method of claim 1 wherein the subject has low testosterone or testosterone

deficiency prior to commencing therapy.

5. The method of claim 1 wherein said subject has at least one or more biomarkers of a liver disease or condition in the upper normal range or above.

6. The method of claim 1 wherein said subject has at least two biomarkers of a liver disease or condition in the upper normal range or above.

7. The method of claim 1 wherein said subject has at least three biomarkers of a liver disease or condition in the upper normal range or above.

8. The method of claim 1 wherein said subject has at least four biomarkers of a liver disease or condition in the upper normal range or above.

9. The method of claim 1 wherein said subject has at least one or more biomarkers of a liver disease or condition elevated above the upper normal limit.

10. The method of claim 1 wherein said subject has at least two biomarkers of a liver disease or condition elevated above the upper normal limit.

11. The method of claim 1 wherein said subject has at least three biomarkers of a liver disease or condition elevated above the upper normal limit.

12. The method of claim 1 wherein said method reduces the level of said subject’s serum alkaline phosphatase levels compared to said subjects baseline value.

13. The method of claim 1 said treatment prolonging the amount of time the subject can wait for a liver transplant or survive.

14. The method of claim 1 wherein the amount of testosterone ester is from 10 mg to 5000 mg per day.

15. The method of claim 1 wherein the testosterone ester is chosen from testosterone undecanoate, testosterone tridecanoate, testosterone dodecanoate, or a combination thereof.

16. The method of claim 1, wherein the pharmaceutical composition comprises a vitamin E, a vitamin E prodrug, a vitamin E derivative, or an omega-3 fatty acid.

17. The method of claim any one of claims 5 to 11 wherein the one or more biomarkers are chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers.

18. The method of any one of claims 1-17 wherein the liver disease or condition is

chosen from hepatitis (all types, including non-alcoholic steatohepatitis (NASH)), cirrhosis (all types), fatty liver disease, including non-alcoholic fatty liver disease (NAFLD), including cirrhosis in a patient at risk, primary NASH or NASH secondary to liver transplantation.

19. The method of any of claims 1-18 wherein said subject in need of treatment has one, two, or three, or more co-morbid diseases or conditions chosen from obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, and cachexia.

20. The method of claim 1 wherein said subject in need of treatment has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH , NAFLD , primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, and an autoimmune disease related to the liver, or has a biomarker level related to or indicative of one or more of these disease or conditions.

21. The method of claim 1 comprising oral administration of 200 mg to 750 mg of

testosterone undecanoate per day to said subject wherein the subject is an adult male subject.

22. The method of claim 1 comprising oral administration of 300 mg to 600 mg of

testosterone undecanoate per day to said subject wherein the subject is an adult male subject.

23. The method of claim 1 comprising oral administration of 450 mg of testosterone undecanoate per day to said subject wherein the subject is an adult male subject.

24. The method of claim 1 comprising oral administration of 400 mg to 2000 mg of testosterone tridecanoate per day to said subject wherein the subject is an adult male subject.

25. The method of claim 1 comprising oral administration of 600 mg to 1800 mg of

testosterone tridecanoate per day to said subject wherein the subject is an adult male subject.

26. The method of claim 1 comprising oral administration of 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to said subject wherein the subject is an adult male subject.

27. The method of any one of claims 21-26 wherein the subject is a female subject

instead of a male subject and the dose of testosterone undecanoate or testosterone tridecanoate is about 1/10th - 1/15th of the value.

28. The method of claim 1 wherein said subject has an age chosen from 1 to 18 years old, 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old, or older than 65 years old.

29. The method of claim 1 wherein said subject has NASH with fibrosis.

30. The method of claim 1 wherein said subject has NASH with F2 or F3 fibrosis (NASH CRN scale).

31. The method of claim 1 comprising oral administration of 100 mg to 200 mg of

testosterone undecanoate per day to said subject wherein the subject is an adult male subject.

32. The method of claim 1 comprising oral administration of 200 mg to 400 mg of testosterone undecanoate per day to said subject wherein the subject is an adult male subject.

33. The method of claim 1 comprising oral administration of 5 mg to 100 mg of

testosterone undecanoate per day to said subject wherein the subject is an adult male subject.

34. The method of claim 1 further comprising treating said subject with one or more additional pharmaceuticals agents suitable for treating liver disease, a comorbidity of liver disease, or symptoms thereof.

Description:
LIVER DISEASE

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of United States Provisional Patent Application Serial Nos. 62/701,309, filed on July 20, 2018; 62/714,968 filed on August 6, 2018; 62/728,580 filed on September 7, 2018; 62/783,190, filed December 20, 2018; and 62/793,724, filed on January 17, 2019, each of which is incorporated herein by reference.

FIELD OF THE INVENTION

This disclosure relates to compositions and methods for treating liver disease and reducing mortality in specific patient populations.

BACKGROUND

Historically, some androgens like anabolic-androgenic compounds were considered to have toxic effects on the liver. See e.g., Bond et al. PMID: 27372877; Neri et al. PMID:

21443508; and Kemp CJ et al. PMID: 2798421.

Despite evidence of hepatotoxicity of androgens in the literature, studies have examined the effect of testosterone on liver fat reduction in fatty liver disease and have general reported inconclusive or negative results in humans. For example, Huang et al. (PMID: 23292288) concluded“Testosterone administration in older men with mobility limitation and low testosterone levels was not associated with a reduction in hepatic fat.” Moreover, elevated testosterone levels in females can associated with fatty liver, liver disease and related comorbidities. Magnussen et al PMID: 28522646 reported“hepatic fat content and VAT were unchanged” in males with type 2 diabetes and low testosterone levels that were treated for 24- weeks with a testosterone gel. Sattler et al PMID: 25392748 also reported that testosterone replacement did not have an effect of liver fat in 20 men with lower baseline levels of testosterone when treated for 20 weeks with a testosterone gel.

There is a need for testosterone/androgen based therapies for specific populations of individuals. More specifically, there is a need for new therapies for liver disease. SUMMARY

Described herein are methods and composition for use in treating, preventing, slowing the progression, or reducing the risk of liver disease (or comorbidities thereof) or symptoms thereof. In particular, it was discovered that androgen receptor modulators e.g., agonists are useful for improving liver function, liver diseases or conditions and related comorbidities. While a number of modalities for treating liver disease are described herein in the context of oral androgen therapy e.g., oral administration of testosterone esters like testosterone undecanoate, the skilled artisan recognizes in view of this disclosure, other androgen receptor agonist can be adapted and employed in these methods such as transdermal, nasal, buccal, and injectable testosterone products. In addition to testosterone, testosterone esters and the such, other androgen receptor agonists may be employed herein as well as methods and compounds, pharmaceuticals, nutritional or vitamin supplements that increase serum testosterone levels, serum free testosterone levels, or androgen receptor signaling in target tissues or compartments.

It was surprisingly discovered that oral testosterone (or androgen) therapy e.g., oral administration of a pharmaceutical composition containing a testosterone ester (e.g., or an androgen receptor agonist or anabolic agent), is particularly useful for treating liver disease. For example, it was found that the instant methods and compositions reduce relevant biomarker levels in patients having elevated biomarkers related to liver disease (e.g., fatty liver disease, liver fibrosis, alcoholic liver disease, hepatitis, steatosis, NAFLD, NASH, NASH with cirrhosis, and comorbidities of testosterone deficiency). Unexpectedly, the methods and compositions disclosed herein, demonstrated a reduction in serum alkaline phosphatase levels that was significantly better than that observed with a once a day topical testosterone product (e.g, AndroGel™ ), a marketed testosterone replacement therapy administered via the transdermal route. Other unexpected findings include substantial reductions (or improvements) in triglyceride levels, biomarkers of liver injury, and biomarkers for other diseases and conditions (e.g., lipoprotein-associated phospholipase A2), a biomarker of cardiovascular disease. Thus, in one implementation, the methods described herein increase the ratio of serum testosterone levels to alkaline phosphatase (e.g., by 5%, 10%, 15%, 20%, or 25% or more). In another

implementation the methods described herein increase the ratio of serum testosterone levels to serum triglyceride levels (e.g., by 5%, 10%, 15%, 20%, or 25% or more). In other implementations, the methods and compositions disclosed herein, improve diseases and conditions (or symptoms or related biomarkers) in conditions that are co-morbid with low testosterone levels (e.g., either total testosterone or free testosterone).

Without wishing to be bound by theory, it is believed that in some contexts, as illustrated herein, testosterone deficient subjects with specific biomarkers in the high normal range or above normal are particularly sensitive to the methods and compositions described herein and see surprising reductions or improvements in the levels of these biomarkers. Again, in many cases these biomarker improvements are both over baseline values and in comparison to an active control (transdermal testosterone preparation). Moreover, populations of individuals that are at higher risk for developing specific diseases and conditions are particularly amenable to the treatments described herein (including those that do not have testosterone deficiency). Thus, the methods and compositions described herein improve or ameliorate biomarkers associated with diseases by 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, or 70% or more compared to baseline values. It is also believed that the method and compositions described herein have utility and can be used in subjects that do not have testosterone deficiency.

It was unexpectedly found that normalization or improvement of biomarkers (e.g., one or more biomarkers) can occur relatively quickly after commencement of the methods using the compositions described herein. For example, treatment for 1, 2, 3, 4, 5, 6, or 7 or more weeks can provide substantial improvements in one or more biomarkers associated with liver disease or mortality. Moreover, the methods and compositions provide sustained responses e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 or 12 months or more. As illustrated in the Examples, treatment with the methods and compositions described herein for 3 weeks can result in substantial improvements of biomarker levels. Typically, the treatments described herein are once-a-day or twice-a-day dosing regimens, although other dosing regimens are contemplated and have been shown to be effective e.g., three times a day. It is also notable that both testosterone undecanoate based oral dosing regimens and testosterone tridecanoate dosing regimens are effective in these treatments. Thus, without wishing to be bound by theory, it is believed that the methods and compositions described herein can be used with numerous testosterone esters and other steroid (and steroid ester containing compositions). For example, the methods and compositions described herein can be used with testosterone esters wherein the ester moiety can have from 2-20 carbons (e.g., the ester moiety is derived from a 2 carbon to 20 carbon fatty acid or fatty acid derivative including straight chain, branched chain, cyclic (e.g., cypionate or cypionic acid), saturated, and unsaturated (oleate or oleic acid) versions of 2 carbon to 20 carbon alkanoic acids). Thus, the compositions including two or more testosterone esters (e.g., testosterone undecanoate and testosterone tridecanoate) can be useful in the methods disclosed herein and have unexpected effects.

Provided herein are methods and compositions useful for treating conditions associated with increased mortality such as liver disease and particularly fatty liver disease, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, steatosis, cirrhosis, alcoholic liver disease and other liver diseases. Moreover, provided herein are methods for treating liver disease or a symptom thereof. Furthermore, provided herein are compositions and methods for modulating biomarkers associated with liver disease or symptoms thereof.

Accordingly, in one embodiment, a method of treating a liver disease or condition is provided, said method comprising identifying a subject in need of treatment e.g., has a liver disease or condition. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject (e.g., in need of treatment). In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect, the subject is from 1 to 18 years old, 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, said subject (e.g., in need of treatment) has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH (“Non- Alcoholic SteatoHepatitis”), NAFLD (“Non-Alcoholic Fatty Liver Disease”), primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, an autoimmune disease related to the liver (or has a biomarker level related to or indicative of one or more of these diseases). In one aspect, said method comprises identifying a subject in need of treatment. In one aspect, the disease or condition is a comorbidity of testosterone deficiency. In a specific aspect, the subject in need of treatment is treated with one or more additional therapeutic agents (e.g., combination therapy). Additionally, this paragraph is to be read as being specifically incorporated into each specific method or method embodiment described in the Detailed

Description.

Accordingly, in one embodiment, a method of treating a liver disease or condition is provided, said method comprising identifying a subject with a biomarker associated with said liver disease or condition where the biomarker is in the upper normal range and orally

administering to said subject a pharmaceutical composition comprising a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) in an amount sufficient to reduce the level of the biomarker or reduce the rate of increase of the biomarker. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL for a male subject or 1/10 th of these values for a female subject. In one aspect, said method comprise oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprise oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg,

750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 & - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, said subject (e.g., in need of treatment) has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH , NAFLD , primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, an autoimmune disease related to the liver (or has a biomarker level related to or indicative of one or more of these diseases). In one aspect, said method comprises identifying a subject in need of treatment. In one aspect, the disease or condition is a comorbidity of testosterone deficiency. In a specific aspect, the subject in need of treatment is treated with one or more additional therapeutic agents (e.g., combination therapy). Additionally, this paragraph is to be read as being specifically incorporated into each specific method or method embodiment described in the Detailed Description.

Accordingly, in one embodiment, a method of treating a liver disease or condition is provided, said method comprising identifying a subject with a biomarker associated with said disease or condition where the biomarker is outside of the normal range (e.g., above or below) and orally administering to said subject a pharmaceutical composition comprising a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) in an amount sufficient to ameliorate or improve the level of the biomarker (e.g., move the level in the direction of the normal range), reduce the rate of increase or decrease of the biomarker, or treat the disease or condition. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL for a male subject or 1/10 th of these values for a female subject. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, said subject (e.g., in need of treatment) has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH, NAFLD, primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, an autoimmune disease related to the liver (or has a biomarker level related to or indicative of one or more of these diseases). In one aspect, said method comprises identifying a subject in need of treatment. In one aspect, the disease or condition is a comorbidity of testosterone deficiency. In a specific aspect, the subject in need of treatment is treated with one or more additional therapeutic agents (e.g., combination therapy). Additionally, this paragraph is to be read as being specifically incorporated into each specific method or method embodiment described in the Detailed

Description.

Accordingly, in yet another embodiment, a method of treating a liver disease or condition is provided, said method comprising identifying a subject with a biomarker associated with said disease or condition where the biomarker is above the upper normal limit and orally

administering to said subject a pharmaceutical composition comprising a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) in an amount sufficient to reduce the level of the biomarker or reduce the rate of increase of the biomarker. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL for a male subject or 1/10 th of these values for a female subject. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg,

750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect, the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, said subject (e.g., in need of treatment) has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH, NAFLD, primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, an autoimmune disease related to the liver (or has a biomarker level related to or indicative of one or more of these diseases). In one aspect, said method comprises identifying a subject in need of treatment. In one aspect, the disease or condition is a comorbidity of testosterone deficiency. In a specific aspect, the subject in need of treatment is treated with one or more additional therapeutic agents (e.g., combination therapy). Additionally, this paragraph is to be read as being specific incorporated into each specific method or method embodiment described in the Detailed Description.

Accordingly, in one embodiment, a method of treating a disease or condition is provided, said method comprising identifying a subject with a biomarker associated with said disease or condition where the biomarker is above twice the upper normal range limit and orally

administering to said subject a pharmaceutical composition comprising a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) in an amount sufficient to reduce the level of the biomarker or reduce the rate of increase of the biomarker. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL for a male subject or l/10th of these values for a female subject. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect, the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, said subject (e.g., in need of treatment) has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH, NAFLD, primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, an autoimmune disease related to the liver (or has a biomarker level related to or indicative of one or more of these diseases). In one aspect, said method comprises identifying a subject in need of treatment. In one aspect, the disease or condition is a comorbidity of testosterone deficiency. In a specific aspect, the subject in need of treatment is treated with one or more additional therapeutic agents (e.g., combination therapy). Additionally, this paragraph is to be read as being specifically incorporated into each specific method or method embodiment described in the Detailed Description.

Accordingly, in one embodiment, a method of treating a liver disease or condition is provided said method comprising identifying a subject with a biomarker associated with said disease or condition where the biomarker is above 3 times the upper normal limit and orally administering to said subject a pharmaceutical composition comprising a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) in an amount sufficient to reduce the level of the biomarker or reduce the rate of increase of the biomarker. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL for a male subject or 1/10 th of these values for a female subject. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprise oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg,

750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, said subject (e.g., in need of treatment) has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH, NAFLD, primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, an autoimmune disease related to the liver (or has a biomarker level related to or indicative of one or more of these diseases). In one aspect, said method comprises identifying a subject in need of treatment. In one aspect, the disease or condition is a comorbidity of testosterone deficiency. In a specific aspect, the subject in need of treatment is treated with one or more additional therapeutic agents (e.g., combination therapy). Additionally, this paragraph is to be read as being specifically incorporated into each specific method or method embodiment described in the Detailed Description.

Accordingly, in one embodiment, a method of treating a liver disease or condition is provided, said method comprising identifying a subject with a biomarker associated with said disease or condition where the biomarker is between the upper normal limit and twice or thrice the upper normal limit, and orally administering to said subject a pharmaceutical composition comprising a testosterone ester (e.g., testosterone imdecanoate or testosterone tridecanoate) in an amoimt sufficient to reduce the level of the biomarker or reduce the rate of increase of the biomarker. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL for a male subject or 1/10 th of these values for a female subject. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone imdecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone imdecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone imdecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, said subject (e.g., in need of treatment) has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH, NAFLD, primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, an autoimmune disease related to the liver (or has a biomarker level related to or indicative of one or more of these diseases). In one aspect, said method comprises identifying a subject in need of treatment. In one aspect, the disease or condition is a comorbidity of testosterone deficiency. In a specific aspect, the subject in need of treatment is treated with one or more additional therapeutic agents (e.g., combination therapy). Additionally, this paragraph is to be read as being specific incorporated into each specific method or method embodiment described in the Detailed Description. Accordingly, in one embodiment, a method of treating a liver disease or condition is provided said method comprising identifying a subject with a biomarker that is greater than the upper normal limit, greater than 2 times the upper normal limit, greater than 3 times the upper normal limit, greater than 4 times the upper limit, or defined by any two of these bounds (e.g., 3- 4 times upper normal limit) associated with said disease or condition and orally administering to said subject a pharmaceutical composition comprising a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) in an amount sufficient to reduce the level of the biomarker or reduce the rate of increase of the biomarker. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL for a male subject or 1/10 th of these values for a female subject. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, said subject (e.g., in need of treatment) has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH, NAFLD, primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, an autoimmune disease related to the liver (or has a biomarker level related to or indicative of one or more of these diseases). In one aspect, said method comprises identifying a subject in need of treatment. In a specific aspect, the subject in need of treatment is treated with one or more additional therapeutic agents (e.g., combination therapy). Additionally, this paragraph is to be read as being specific incorporated into each specific method or method embodiment described in the Detailed Description.

Accordingly, in one embodiment, a method of treating a liver disease or condition is provided, said method comprising identifying a subject with a biomarker associated with said liver disease or condition where the biomarker is an imaging biomarker (e.g., liver) or a liver histology biomarker and orally administering to said subject a pharmaceutical composition comprising a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) in an amount sufficient to reduce the level of the biomarker or reduce the rate of increase of the biomarker. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL for a male subject or 1/10th of these values for a female subject. In one aspect, said method comprise oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprise oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10th - 1/15th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56- 65 years old or older than 65 years old. In one aspect, said subject (e.g., in need of treatment) has a disease or condition chosen from metabolic syndrome, diabetes (e.g., Type 2 Diabetes), hypertension, obesity, hypogonadism, hypertriglyceridemia, fatty liver disease, liver fibrosis, steatosis, cirrhosis, hepatitis, cardiovascular disease, NASH , NAFLD, primary sclerosing cholangitis, is waiting for a liver transplant, has received a liver transplant, primary biliary cholangitis, a liver disease or condition associated with chronic inflammation, hepatocellular carcinoma, graft versus host disease e.g., related to liver transplant, an autoimmune disease related to the liver (or has a biomarker level related to or indicative of one or more of these diseases). In one aspect, said method comprises identifying a subject in need of treatment. In one aspect, the disease or condition is a comorbidity of testosterone deficiency. In a specific aspect, the subject in need of treatment is treated with one or more additional therapeutic agents (e.g., combination therapy). Additionally, this paragraph is to be read as being specifically incorporated into each specific method or method embodiment described in the Detailed Description.

In some aspects of the methods described above and elsewhere in this disclosure, male subjects having serum testosterone levels in the range of less than 400 ng/dL, less than 450 ng/dL, less than 500 ng/dL, less than 550 ng/dL, less than 600 ng/dL, or less than 700 ng/dL can be treated with the compositions described herein in accordance with methodology described herein.

In some aspects of the methods described above and elsewhere in this disclosure, male subjects having serum testosterone levels in the range of 50-100 ng/dL, 100-150 ng/dL, 150-200 ng/dL, 200-250 ng/dL, 250-300 ng/dL, 300-350 ng/dL, 350-400 ng/dL, 400-450 ng/dL, 450-500 ng/dL, 550-600 ng/dL, 600-700 ng/dL, or any combination of these ranges can be treated with the compositions described herein in accordance with methodology described herein.

In some aspects of the methods described above and elsewhere in this disclosure, male subjects having serum SHBG levels in the range of less than 20 nmol/L, less than 30 nmol/L, less than 40 nmol/L, less than 50 nmol/L, less than 60 nmol/L, less than 70 nmol/L, less than 80 nmol/L, less than 90 nmol/L, less than 100 nmol/L, less than 150 nmol/L, less than 200 nmol/L, less than 300 nmol/L, less than 400 nmol/L, or e.g., less than 1000 ng/dL can be treated with the compositions described herein in accordance with methodology described herein. These serum SHBG ranges can be used in conjunction with the serum testosterone levels described herein for specific populations particularly suited to treatment as described herein.

In some aspects of the methods described above and elsewhere in this disclosure, male subjects having serum testosterone levels in the range of 1-20 nmol/L, 20-25 nmol/L, 25-30 nmol/L, 30-35 nmol/L, 35-40 nmol/L, 40-45 nmol/L, 45-50 nmol/L, 50-55 nmol/L, 55-60 nmol/L, 60-65 nmol/L, 65-80 nmol/L, 80-100 nmol/L, 100-125 nmol/L, 125-150 nmol/L, 150- 200 nmol/L, 200-300 nmol/L, 300-450 nmol/L, or e.g., 450-750 nmol/L, or any combination of these ranges can be treated with the compositions described herein in accordance with methodology described herein. These serum SHBG ranges can be used in conjunction with the serum testosterone levels described herein for specific populations particularly suited to treatment as described herein.

In one aspect of each of the methods described herein (e.g., each paragraph above in the Summary and each method embodiment of the Detailed Description), the method increases the serum testosterone level of the subject. In one aspect, the method increases the serum

testosterone level (e.g., Caveo-24) of the subject by at least 10, 20, 30, 40, or 50 ng/dL over the subject’s baseline (pre-treatment) value (e.g., for a male subject or adjusted accordingly for female or pediatric subjects). In one aspect, the method increases the serum testosterone level (e.g., Caveo-24) of the subject by at least 30, 50, 75, 100, 150, 200, 300, 400 or 500 ng/dL over the subject’s baseline (pre-treatment) value (e.g., for a male subject or adjusted accordingly for female or pediatric subjects). The baseline serum testosterone level for the subject can be determined by a single morning blood draw. Alternatively, the baseline serum testosterone level can be determined by the average of two morning blood draws from different days. The increase over the baseline value can be estimated as a Cave serum testosterone level in a manner deemed appropriate by a relevant medical professional. For example, the Cave serum testosterone level can be estimated as Caveo-24, Caveo-12, Caveo-e, Caveo-6, Caveo-4, Caveo-3 and Caveo-2. Alternatively, a clinician can ascertain that a sufficient increase in serum testosterone level has occurred via the observation of clinical outcomes or characteristics (e.g., improvement in symptoms), or modulation of one or more biomarkers.

In another aspect of each of the methods described herein (e.g., each paragraph above in the Summary and each method embodiment of the Detailed Description), the method produces a serum testosterone profile over a 24-hour period that approximates the pattern of a healthy young subject (not necessarily the absolute levels). That is to say there is an increase in serum testosterone levels in the morning which tapers off during the remainder of the day. Thus, in one aspect the 24-hour serum testosterone profile is characterized as substantially different than that provided by other means of testosterone therapy. For example, commonly used and prescribed testosterone therapies include transdermal testosterone therapy which over the course of a 24- hour period produces a relative flat pharmacokinetic profile or injectable testosterone therapies (e.g., intramuscular or subcutaneously) which provide relatively flat pharmacokinetic profiles over a 24-hour period and longer, depending on the specific therapy. Another way of characterizing this difference is the difference or delta between serum testosterone Cmax and serum testosterone Caveo-24. With both transdermal and injectable therapy the difference between serum testosterone Cmax and serum testosterone Caveo-24 is relatively small compared to that of orally administered testosterone therapies. Thus, in some aspects, the methods and compositions described herein provide Cmax to Cave ratios (e.g., Cmax/Caveo-24) of greater than 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9 or 2.0. Thus, in some aspects, the methods and compositions described herein provide Cmax to Cave ratios (e.g., Cmax/Caveo-24) of less than 100, 90, 80, 70, 60, 50, 40, 35, 30 or 25. In some aspects, the ratio of Cmax/Caveo-24 is defined as a range defined by any two values in the previous two sentences.

In one embodiment, the methods described above can include the use of an injectable or transdermal testosterone therapy in conjunction with an oral therapy. For example, the injectable or transdermal therapy can increase the subject’s basal serum testosterone level and the oral therapy can be used to provide a 24-hour serum testosterone profile that mimics a normal physiological profile. Alternatively, the methods described herein can be adapted for use with non-oral testosterone therapies including but not limited to (1) injectable testosterone products e.g., testosterone enanthate, testosterone propionate, testosterone undecanoate, or another testosterone ester (2) transdermal or topical testosterone products like Androgel, Testim, or Axiron, buccal testosterone products, (3) nasal testosterone products or any other androgen based therapy (e.g., androgen receptor agonist). In a specific aspect, these non-oral products can be used to target the specific disease states, populations, biomarkers and the such as described for the oral compositions.

In one embodiment, the methods described herein involve oral administration of a testosterone ester (e.g., testosterone undecanoate, testosterone tridecanoate, or a combination thereof) formulated with vitamin E, a vitamin E prodrug, or a vitamin E derivative. In a preferred aspect, the Vitamin E compound is d-alpha-tocopherol or d-alpha tocopherol acetate. In another preferred aspect, the amount of d-alpha-tocopherol (or it’s acetate) administered per day is from about 100 IU to about 2000 IU, 200 IU to about 1600 IU per day, about 400 IU to about 1000 IU per day or about 600 IU to 900 IU per day. These amounts can be co-formulated with the testosterone esters or administered separately. The exemplary formulations described herein can be adapted accordingly to use d-alpha-tocopherol (or e.g., a prodrug thereof (d-alpha- tocopheryl acetate)), tocotrienol and other related Vitamin E related compounds.

Without wishing to be bound by theory, it is believed the compositions and method described herein are useful and can be used to treat steatosis (or fibrosis or symptoms thereof) in any tissue or cell, including, but not limited to, one or more one or more of the following:

pancreatic steatosis, renal steatosis, cardiac steatosis, pulmonary steatosis, intestinal steatosis, cerebral steatosis, and muscular steatosis, or a symptom thereof. In some aspects, the cell or tissues expresses androgen receptor.

Without wishing to be bound by theory, it is believed the compositions and method described herein are useful and can be used to treat macrovesicular steatosis or microvesicular steatosis.

Male hypogonadism (testosterone, T, level < 300 ng/dL) is an underappreciated comorbidity in non-alcoholic fatty liver disease (NAFLD)/ steatohepatitis (NASH). In addition to the well-established link between low T levels and facets of the metabolic syndrome (obesity, hypertension, hypertriglyceridemia, and type 2 diabetes), hypogonadism is also associated with NAFLD/NASH in males.

This disclosure relates to the prevalence of fatty liver in hypogonadal patients, as identified by Magnetic Resonance Imaging-Proton Density Fat Fraction (MRI-PDFF), a non- invasive quantitative biomarker of liver fat content.

In an a prospective design open-label, multi-center, single arm‘Liver Fat Study’ evaluating LPCN 1144 (oral testosterone undecanoate) treatment in hypogonadal patients, baseline % liver fat was assessed via MRI-PDFF in a subset of study patients (N=36).

The prevalence of features of the metabolic syndrome in the study population was: 81% obese, 58% hypertriglyceridemia, 28% hypertensive, and 17% type 2 diabetes. Body Mass Index (BMI), T level, and liver fat fraction were 34±6 kg/m2, 199±61 ng/dL, and 8.8±7.9% (mean±SD), respectively. Based on liver fat > 5%, about 70 % of hypogonadal patients were identified as having NAFLD. Among hypogonadal patients with NAFLD, obesity was the most prevalent comorbidity (88%). Moreover, the prevalence ofNAFLD increased in hypogonadal patients with increasing BMI, with the prevalence rising to 100% with Class IP obesity (BMI > 40 kg/m2).

In conclusion, a high prevalence ofNAFLD was observed in this Liver Fat Study.

Among the evaluated comorbidities, obesity has the strongest association with NAFLD in the hypogonadal male patients. Thus provided herein is a method of treating a subject in need of treatment, said method comprising determining (1) the subject’s serum or plasma testosterone level; (2) the subject’s BMI, wherein a subject having (A) low testosterone or is hypogonadal and (B) is obese or has a BMI of 30 kg/m 2 or greater, is treated for fatty liver disease. In another implementation, a method of diagnosing a subject is provided, said method comprising determining (1) the subject’s serum or plasma testosterone level; (2) the subject’s BMI, wherein a subject having (A) low testosterone or is hypogonadal and (B) is obese or has a BMI of 30 kg/m 2 or greater, is likely to have fatty liver disease and is assessed by a liver imaging technique or has one or more biomarkers assessed to determine the stage of fatty liver disease. The method can further comprise administering to said subject a pharmaceutical agent for treating NAFLD or NASH.

Thus, also provided herein are methods for diagnosing, staging, prognosing and treating subj ects having fatty liver disease.

It is to be understood that the methods described in this section are applicable to the methods described for the specific conditions/diseases in the Detailed Description.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1 shows a summary of an analysis of liver function enzyme changes from baseline to end of study values comparing obese (e.g., BMI> 30 kg/m 2 ) to non-obese subjects in the study of Example 1.

Figure 2 shows a summary of an analysis of serum alanine transaminase levels of subjects in the study of Example 1, comparing baseline values to end of study values. In this figure and the remaining figures, Q-max refers to the maximum value of the biomarker for the specified quartile. For example, in this figure the first quartile are those subjects having serum alanine transaminase levels of less than or equal to 21 U/L, the second quartile are those subjects having serum alanine transaminase levels between 21 U/L and less than or equal to 27 U/L.

Figure 3 shows a summary of an analysis of serum alanine transaminase levels of subjects in the highest quartile of alanine transaminase levels at baseline during the course of the study of Example 1.

Figure 4 shows a summary of an analysis of serum aspartate transaminase levels of subjects comparing baseline to end of study values in the study of Example 1.

Figure 5 shows a summary of an analysis of serum alkaline phosphatase levels of subjects comparing baseline to end of study values in the study of Example 1.

Figure 6 shows a summary of analyses of serum alkaline phosphatase levels of subjects comparing baseline to end of study values in the studies of Examples 1, 2, 3, and 4.

Figure 7 shows a summary of an analysis of serum gamma-glutamyl transferase levels of subjects comparing baseline to end of study values in the study of Example 1.

Figure 8 shows a summary of an analysis of serum bilirubin levels of subjects comparing baseline to end of study values in the study of Example 1.

Figure 9 shows a summary of an analysis of serum triglyceride levels of diabetic and non-diabetic subjects at baseline compared to end of study values in the study of Example 1.

Figure 10 shows a summary of an analysis of serum triglyceride levels of subjects having lipid metabolism disorder and non-lipid metabolism disorder subjects at baseline compared to end of study values in the study of Example 1.

Figure 11 shows a summary of an analysis of serum triglyceride levels of subjects at baseline compared to end of study in the study of Example 1.

Figure 12 shows a summary of an analysis of serum triglyceride levels of subjects at baseline compared to end of study in the study of Examples 1, 2, and 4. Figure 13 shows a summary of an analysis of serum LDL levels of subjects at baseline compared to end of study in the study of Example 1.

Figure 14 shows a summary of an analysis of serum total cholesterol levels of subjects at baseline compared to end of study in the study of Example 1.

Figure 15 shows a summary of an analysis of serum non-HDL cholesterol levels of subjects at baseline compared to end of study in the study of Example 1.

Figure 16 illustrates the difference in serum testosterone levels for oral testosterone therapy (results from Example 2) and transdermal therapy (which is also similar to injectable therapies in the sense of a relatively flat profile).

Figure 17 shows the prevalence of comorbidities in the male hypogonadal population studied herein.

Figure 18 shows the mean liver fat % as a function of BMI classes in the male hypogonadal population studied herein.

Figure 19 shows percent of subjects having NAFLD as defined by liver fat cut-off values of 5% and 8%, as a function of BMI class, for male hypogonadal population studied herein.

Figure 20 the distribution of liver fat % values across the male hypogonadal population studied herein.

Figure 21 shows the prevalence of comorbidities in the male hypogonadal population studied herein according to liver fat % cut-offs.

The Figures illustrate specific aspects of the compositions and methods for using such compositions. Together with the following description, the Figures demonstrate and explain the principles of the methods and compositions produced through these methods. In the drawings, the thickness of layers and regions are exaggerated for clarity. Also, directions (e.g., above, below, top, bottom, side, up, down, under, over, upper, lower, horizontal, vertical, "x," "y," "z," etc.), if provided, are relative and provided solely by way of example and for ease of illustration and discussion and not by way of limitation. In addition, where reference is made to a list of elements (e.g., elements a, b, c), such reference is intended to include any one of the listed elements by itself, any combination of less than all of the listed elements, and/or a combination of all of the listed elements.

DETAILED DESCRIPTION

It was unexpectedly found that androgen receptor modulators e.g., agonists are useful for improving liver function, liver diseases or conditions and related comorbidities. While a number of modalities for treating liver disease are described herein in the context of oral androgen therapy e.g., oral administration of testosterone esters like testosterone undecanoate, the skilled artisan recognizes in view of this disclosure, other androgen receptor agonist can be adapted and employed in these methods such as transdermal, nasal, buccal, and injectable testosterone products. In addition to testosterone, testosterone esters and the such, other androgen receptor agonists (including selective androgen receptor modulators, anabolic steroids wtc.) may be employed herein as well as methods and compounds, pharmaceuticals, nutritional or vitamin supplements that increase serum testosterone levels, serum free testosterone levels, or androgen receptor signaling in target tissues or compartments.

The following description supplies specific details in order to provide a thorough understanding of the methods and compositions of the invention. Nevertheless, the skilled artisan would understand that the compositions and associated methods of making and using such compositions can be implemented and used without employing these specific details. Indeed, the compositions and associated methods can be placed into practice by modifying the illustrated devices and methods and can be used in conjunction with any other agents and techniques conventionally used in the industry. For example, while the description refers to specific indications, it could be modified to be used in other indications.

It was surprisingly discovered that oral androgen therapy e.g., oral administration of a pharmaceutical compositions containing a testosterone ester (e.g., or an androgen receptor agonist or anabolic agent), is particularly useful for treating liver disease, comorbidities of testosterone deficiency, and can reduce mortality, and ameliorate or improve biomarkers related to these conditions or diseases. For example, it was found that the instant methods and compositions reduce relevant biomarker levels in patients having elevated biomarkers related to liver disease (e.g., fatty liver disease, liver fibrosis, alcoholic liver disease, hepatitis, steatosis, NAFLD, NASH, NASH with cirrhosis, and comorbidities of testosterone deficiency). Unexpectedly, as described herein, the reduction in serum alkaline phosphatase levels was significantly better with oral therapy than that observed with once a day topical testosterone product (e.g, AndroGel)™, a marketed testosterone replacement therapy administered via the transdermal route. Other unexpected findings include substantial reductions (or improvements) in triglyceride levels, biomarkers of liver injury, and biomarkers for other diseases and conditions associated with lipoprotein-associated phospholipase A2, a biomarker of

cardiovascular disease. Thus, in one implementation, the methods described herein increase the ratio of serum testosterone levels to alkaline phosphatase. Thus, in one implementation, the methods described herein increase the ratio of serum testosterone levels to serum triglycerides.

While not wishing to be bound by theory, it is believed in some aspects described herein, that the methods and compositions present a therapy approximating a normal physiological pattern of serum testosterone levels with peaks and troughs of serum testosterone levels as opposed to the relative flat profile of other testosterone therapies like transdermal products and injectable products. It is believed that, in some aspects, the more normal physiological pattern of serum testosterone levels provides less suppression of endogenous serum testosterone levels and relative hormones/biomarkers leading to the unexpected results described herein.

Concentrations, amounts, levels and other numerical data may be expressed or presented herein in a range format. It is to be understood that such a range format is used merely for convenience and brevity and thus should be interpreted flexibly to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges or decimal units encompassed within that range as if each numerical value and sub-range is explicitly recited. As an illustration, a numerical range of“about 1 to about 5” should be interpreted to include not only the explicitly recited values of about 1 to about 5, but also include individual values and sub-ranges within the indicated range. Thus, included in this numerical range are individual values such as 2, 3, and 4 and sub-ranges such as from 1-3, from 2-4, and from 3-5, etc., as well as 1, 2, 3, 4, and 5, individually. This same principle applies to ranges reciting only one numerical value as a minimum or a maximum. Furthermore, such an interpretation should apply regardless of the breadth of the range or the characteristics being described. The terms“serum testosterone” or“serum (17-P)-Hydroxy-4-Androsten-3-one levels,” “serum T levels,”“serum testosterone concentration,”“plasma testosterone concentration,” “testosterone concentration in the blood,” and“serum testosterone concentration,” are used interchangeably and refer to the“total” testosterone concentration which is the sum of the bioavailable testosterone including free and bound testosterone concentrations. Unless otherwise specified, these values are“observed” testosterone concentrations without adjusting or correcting for the base-line serum testosterone levels in the subject(s). As with any bio-analytical measure, for increased consistency, the method employed to measure initial serum testosterone levels should be consistent with the method used to monitor and re-measure serum testosterone levels during clinical testing and testosterone therapy for a subject. Unless otherwise stated, “testosterone concentration” refers to serum total testosterone concentration.

Average serum testosterone concentrations can be determined using methods and practices known in the art. For example, the average baseline plasma testosterone concentration of a human male is the arithmetic mean of the total plasma testosterone concentration determined on at least two consecutive time points that are reasonably spaced from each other, for example from about 1 hour to about 168 hours apart. In a particular case, the plasma testosterone concentration can be determined on at least two consecutive times that are about 12 hours to about 48 hours apart. In another particular method, the plasma testosterone concentration of the human male can be determined at a time between about 5 o’clock and about 11 o’clock in the morning. Further, the plasma testosterone concentration can be the determined by standard analytical procedures and methods available in the art, such as for example, automated or manual immunoassay methods, liquid chromatography or liquid chromatography- tandem mass spectrometry (LC-MSMS) etc.

As used herein,“free testosterone serum concentration”, refers to the testosterone concentration not bound to protein e.g., SHBG or albumin. In some aspects of the methods described herein, free testosterone serum concentrations are used instead of serum total testosterone concentrations. For example, a subject can appear to have total serum testosterone levels in the normal range but can be still considered hypogonadal or testosterone deficient based on free testosterone levels. As used herein,“in need of treatment” refers to a subject that has a disease or is suspected of having the disease according to various diagnostic criteria typically used in practice, or desires treatment or is indicated for treatment. Thus,“in need of treatment” can include the step of identifying a subject in need of treatment.

As used herein,“identifying a subject in need of treatment” can include the step of obtaining a biological sample from the subject and determining the level of one or more biomarkers as described herein, assessing the histology of a biological sample obtained from said subject, performing an imaging analysis on the subject, assessing one or more clinical characteristics of said subject (e.g., assessing symptoms or overt symptoms), or a combination thereof.

As used herein, the term AUCu-u is the area under the curve of a plasma-versus-time graph determined for the analyte from the time“tl to time t2”. Wherein tl and t2 are times (in hours) post dosing. For Example, tl could be 1 hour and t2 could be 2 hours.

As used herein, the term“Cavg,”“Cave,” or“C-average” are used interchangeably, and is determined as the AUCu-u mean AUC divided by the time period (|tl-t2|). For example, Cavgio-e is the average plasma concentration over a period of 8 hours from tl=0 to t2=8 hours) postdosing determined by dividing the AUC to-ts value by 8. Similarly, Cavgto-m is the average plasma concentration over a period of 12 hours post-dosing determined by dividing the AUCto-ti2 value by 12 (tl=0-t2=12). Similarly, Cavg 112424 is the average plasma concentration over a period of 12 hours post-dosing determined by dividing the AUCti2424 value by 12 (tl=12-t2=24); Cavg424 is the average plasma concentration over a period of 24 hours post-dosing determined by dividing the AUC10424 value by 24 (tl=0-t2=24), and so on. Unless otherwise stated, all Cavg values are considered to be Cavg424 and unless otherwise stated, all the time values are expressed in hours (h). For example, the term Cavg 10424 denotes Cavg from time zero (0) to 24 hours post dosing.

“Androgen receptor agonists” as used herein refers to compounds or molecules such as testosterone that bind and activate the androgen receptor. Androgen receptor agonists include, but are not limited to, dihydrotestosterone, mibolerone, testosterone, methyltrienolone, oxandrolone, nandrolone and fluoxymesterone. Where appropriate, fatty acid esters of these androgen receptor agonists can be used herein accordingly.

“Testosterone ester” as used herein refers to testosterone esterified with a fatty acid and includes but is not limited to, testosterone undecanoate, testosterone tridecanoate, testosterone enanthate, testosterone decanoate, testosterone palmitate, testosterone cypionate, and testosterone propionate. In one aspect, a preferred testosterone ester for oral administration according to the methods and compositions described herein is testosterone undecanoate. In one aspect, a preferred testosterone ester for oral administration according to the methods and compositions described herein is testosterone decanoate. In one aspect, a preferred testosterone ester for oral administration according to the methods and compositions described herein is testosterone dodecanoate. In one aspect, a preferred testosterone ester for oral administration according to the methods and compositions described herein is testosterone tridecanoate. In one aspect, a preferred testosterone ester for oral administration according to the methods and compositions described herein is testosterone tetradecanoate.

“Fibrosis," or“Liver Fibrosis” as used herein, refers to the accumulation of extracellular matrix constituents that occurs following trauma, inflammation, tissue repair, immunological reactions, cellular hyperplasia, and neoplasia.

“Normal Range” as used herein refers to a range of values generally considered to be representative of a healthy individual or population. It is understood that“normal range” is general specific to a type of assay or lab. For example, for a particular analyte, a normal range for one assay or lab can differ as compared to a normal range for another assay or lab for the same analyte. Thus, the normal ranges described herein may vary from lab to lab or assay to assay. The skilled artisan understands that the normal ranges disclosed herein can vary from individual to individual. The skilled artisan understands that the normal ranges described herein are typically appropriate for the indicated sample e.g., serum and that other samples e.g., saliva can have different normal ranges.

“Upper Normal Range” as used herein refers to above the 50% level for that range. For example, a biomarker may have a normal range of 10-40 U/L, according to this definition, the upper normal range is above 25 U/L and below 40 U/L. In another example, consider a biomarker having a range of 43-115 U/L, the high normal range is above 79 U/L and below 115

U/L.

As used herein,“ALT normal range” refers to the range of serum alanine transaminase values considered normal for healthy individuals and is from about 10 - 40 U/L according to the assay/lab used in Example 1.

As used herein,“AST normal range” refers to the range of serum aspartate transaminase values considered normal for healthy individuals and is from about 10 - 43 U/L according to the assay/lab used in Example 1.

As used herein,“ALP normal range” refers to the range of serum alkaline phosphatase values considered normal for healthy individuals and is from about 43 - 115 U/L according to the assay/lab used in Example 1.

As used herein,“GGT normal range” refers to the range of serum gamma-glutamyl transferase values considered normal for healthy individuals and is from about 10 - 49 U/L according to the assay/lab used in Example 1.

As used herein,“triglyceride normal range” refers to the range of serum triglyceride values considered normal for healthy individuals and is from about 45 - 200 mg/dL according to the assay/lab used in Example 1.

As used herein,“LDL normal range” and is from about 50 - 160 mg/dL according to the assay/lab used in Example 1.

As used herein,“desirable total cholesterol” or“total cholesterol normal range” and for adults is from about 125 - 200 mg/dL according to the assay/lab used in Example 1.

As used herein,“non-HDL cholesterol normal range” for adults is 130 - 159 mg/dL (3.4 - 4.0 mmol/L) is considered near ideal. The skilled artisan realizes that an ideal level depends on a number of factors and the normal range is variable depending on these factors.

As used herein,“VLDL” refers to very low density lipoprotein and has a normal range of

2 to 30 mg/dL. As used herein,“SHBG” refers to sex hormone binding globulin and has a normal range of about 20 to 60 nmol/L for healthy adult males. As with any other of the biomarkers disclosed herein the normal ranges depend on a number of factors including sex and age e.g., Adult female, premenopausal 40-120 nmol/L; Adult female, postmenopausal 28-112 nmol/L; Adult male; 20- 60 nmol/L; Infant (1-23 months) 60-252 nmol/L; Prepubertal (2 years-8 years) 72-220 nmol/L;

Pubertal female 36-125 nmol/L; and Pubertal male 16-100 nmol/L.

As used herein, terms such as fatty liver disease, liver fibrosis, alcoholic liver disease, hepatitis, steatosis, NAFLD, NASH, and NASH with cirrhosis are given their customary meaning to one of ordinary skill in the art and such condition are identifiable and diagnosable by medical professionals such as physicians, hepatologists, gastroenterologists and the like based on assessment of the relevant disease characteristics in patients.

Methods

As described below, it was discovered that compositions containing steroids e.g., steroid esters can be used to treat subjects e.g., male or female human subjects with a variety of diseases and conditions including, but not limited to, those described in the summary section herein or as described below. The ordinary skilled artisan understand that these composition and methods can be used for other diseases and conditions based on this disclosure.

Although some of the description focuses on the oral administration of testosterone esters, the methods and compositions described herein can be adapted to other pharmaceuticals such as androgen agonists, androgens, androgenic-anabolic compounds, selective androgen receptor modulators and other compounds that target androgen receptor signaling pathways in view of the studies described herein.

In one embodiment, a method of reducing the risk of steatosis progressing to cirrhosis in a subject is provided said method comprising oral administration of a pharmaceutical

composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment e.g., having steatosis or at risk for having steatosis progressing to cirrhosis. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating an increased risk of steatosis progressing to cirrhosis. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect of this method, oral administration of the testosterone ester reduces the risk of steatosis progressing to cirrhosis while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect of this method, the subject has alcoholic or non-alcoholic steatosis. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect, said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral

administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprise oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In another embodiment, a method of increasing the amount of time a subject having cirrhosis can survive while waiting for a liver transplant is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment e.g., having cirrhosis or at risk of dying from cirrhosis or is waiting for a liver transplant. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating compromised survival time while waiting for a liver transplant. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect of this method, oral administration of the testosterone ester increases the amount of time a subject having cirrhosis can survive while waiting for a liver transplant while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, the treatment reduces the subject’s MELD score or reduces the rate of increase. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect, said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In yet another embodiment, a method of reducing fatty liver in a subject is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment e.g., has fatty liver or fatty liver disease or has increased risk of having fatty liver disease. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating fatty liver or increasing fatty liver. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect of this method, the subject has testosterone deficiency. In one aspect of this method, oral administration of the testosterone ester reduces fatty liver while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of

testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In yet another embodiment, a method of reducing the rate of fatty liver increase in a subject is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment e.g., has fatty liver or increasing fatty liver. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating increasing fatty liver. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester reduces the rate of fatty liver increase while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In one embodiment, a method of reducing progression or the risk of progression of fatty liver to NAFLD in a subject having or suspected of having fatty liver is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment e.g., has fatty liver or is at risk of having fatty liver progressing to NAFLD. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating an increased risk of fatty liver progressing to NAFLD. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect of this method, oral administration of the testosterone ester reduces progression of fatty liver disease to NAFLD while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprise oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg,

750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities:

obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In yet another embodiment, a method of reducing progression or the risk of progression of NAFLD to NASH in a subject having or suspected of having NAFLD is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment e.g., has NAFLD or is at risk of having NAFLD progressing to NASH. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical

characteristics) indicating an increased risk of NAFLD progressing to NASH. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, the one or more

biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect of this method, oral administration of the testosterone ester reduces the risk of steatosis progressing to cirrhosis while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect of this method, oral administration of the testosterone ester reduces the risk of NAFLD progressing to NASH while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In one embodiment, a method of reducing progression or the risk of progression of NASH to cirrhosis in a subject having or suspected of having NASH is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment e.g., has NASH or is at risk of having NASH progressing to cirrhosis. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical

characteristics) indicating an increased risk of NASH progressing to cirrhosis. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester reduces the progression of NASH to cirrhosis while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprise oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities:

obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia. In an embodiment, a method for treating a subject at risk of having steatosis progressing to cirrhosis is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating an increased risk of steatosis progressing to cirrhosis. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester reduces the risk of steatosis progressing to cirrhosis while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject.

In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg,

1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities:

obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In one embodiment, a method for improving survival of a subject waiting for a liver transplant or decreasing mortality is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to a subject in need of treatment. In one aspect, the subject is in need of treatment e.g., is on a liver transplant waiting list or is in need of a liver transplant. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating compromised survival. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester improves the survival (e.g., reduces mortality) of subjects while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprise oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject.

In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg,

1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities:

obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In an embodiment, a method of improving serum alkaline phosphatase levels in a subject is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject in need of treatment (e.g., having liver disease or at risk of having liver disease). In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating a need for reduction in alkaline phosphatase levels. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester improves serum alkaline phosphatase levels while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In another embodiment, a method for improving serum alanine aminotransferase levels in a subject is provided said method comprising oral administration of a pharmaceutical

composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment (e.g., has liver disease or is at risk of having liver disease). In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating a need for improving serum aminotransferase levels. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect of this method, oral administration of the testosterone ester improves serum alanine transaminase levels while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of

testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect, the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia. In yet another embodiment, a method for improving serum aspartate aminotransaminase levels in a subject is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment (has liver disease or is at risk of having liver disease). In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating a need for improving serum aspartate aminotransaminase levels. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2- 3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester reduces the risk of steatosis progressing to cirrhosis while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect, said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 46-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In another embodiment, a method for treating or preventing hepatic steatosis in a subject is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject.

In one aspect, the subject is in need of treatment (e.g., has liver disease or is at risk of having liver disease). In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating an increased risk of hepatic steatosis. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect, said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject.

In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In one embodiment, a method of improving the ratio of serum alkaline phosphatase levels to (a) aspartate aminotransaminase levels, (b) alanine aminotransferase or (c) both (b) and (c) in a subject is provided said method comprising oral administration of a pharmaceutical

composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment (has liver disease or is at risk of having liver disease). In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating a need for improving the ratio of serum alkaline phosphatase levels to (a) aspartate aminotransaminase levels, (b) alanine

aminotransferase or (c) both (b) and (c). In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect of this method, oral administration of the testosterone ester improves serum aspartate transaminase levels while not substantially increasing serum levels of or decreases serum levels of lipoprotein- associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect, said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 46-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In yet another embodiment, a method of treating anemia in a subject is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject in need of treatment (e.g., has liver disease or is at risk of having liver disease). In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect of this method, oral administration of the testosterone ester treats anemia in a subject having liver disease while not substantially increasing serum levels of or decreases serum levels of Lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect, said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg,

750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old,

36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities:

obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In again another embodiment, a method of treating graft rejection in a subject is provided said method comprising oral administration of a pharmaceutical composition having a

testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject (e.g., has received a liver transplant). In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating an increased risk of graft rejection. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral

administration of the testosterone ester treats graft rejection while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprise oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg,

750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities:

obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia. In another embodiment, a method of treating alcoholic steatosis in a subject is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) related to alcoholic steatosis. In one aspect, said subject is in need of treatment e.g., has an increased risk of alcoholic steatosis or progression of alcoholic steatosis. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester treats alcoholic steatosis while not substantially increasing serum levels of or decreases serum levels of Lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In another embodiment, a method of treating alcoholic cirrhosis in a subject is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject (e.g., having alcoholic cirrhosis). In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) related to alcoholic cirrhosis. In one aspect, said subject is in need of treatment e.g., has an increased risk of alcoholic cirrhosis or progression of alcoholic cirrhosis or increased risk of death from alcoholic cirrhosis. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester treats alcoholic cirrhosis while not substantially increasing serum levels of or decreases serum levels of

Lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In another embodiment, a method of treating alcoholic liver disease in a subject is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) of alcoholic liver disease. In one aspect, said subject is in need of treatment e.g., has an increased risk of alcoholic liver disease or progression of alcoholic liver disease or increased risk of death from alcoholic liver disease. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester treats alcoholic liver disease while not substantially increasing serum levels of or decreases serum levels of Lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of

testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10 th - 1/15 th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In one aspect, the methods described herein are useful for treating a fibrotic condition of the liver. In certain embodiments, the fibrotic condition of the liver is chosen from one or more of: fatty liver disease, steatosis (e.g., nonalcoholic steatohepatitis (NASH), cholestatic liver disease (e.g., primary biliary cirrhosis (PBC), cirrhosis, alcohol-induced liver fibrosis, biliary duct injury, biliary fibrosis, cholestasis or cholangiopathies. In some embodiments, hepatic or liver fibrosis includes, but is not limited to, hepatic fibrosis associated with alcoholism, viral infection, e.g., hepatitis (e.g., hepatitis C, B or D), autoimmune hepatitis, non-alcoholic fatty liver disease (NAFLD), progressive massive fibrosis, exposure to toxins or irritants (e.g., alcohol, pharmaceutical drugs and environmental toxins).

The methods and compositions described herein are also directed to methods of inhibiting or reducing the likelihood of liver injury in a patient at risk for same occurring from or secondary to a variety of etiologies especially including hepatitis (all forms, especially including hepatitis viral), non-alcoholic fatty liver diseases (NAFLD), including non-alcoholic steatohepatitis (NASH), NAFLD or NASH including primary NASH, NASH secondary to liver transplantation (NASH post-liver transplantation), preservation injury of donated organs, acute and chronic liver transplant rejection and metabolic conditions including, for example, Wilson's disease, hemochromatosis, and alpha one antitrypsin deficiency represent alternative aspects of the present invention. In this method, an effective amount of a composition described herein is administered to a patient at risk for liver injury as described above in order to inhibit or reduce the likelihood of liver injury as described above. As a consequence of the actions of compounds according to the present invention in reducing and/or inhibiting liver injury, certain

complications of liver injury may be reduced including, for example, liver failure, liver shock, obstructive jaundice, cirrhosis, including primary biliary cirrhosis, primary sclerosing cholangitis, portal hypertension, ascites, variceal bleeding, encephalopathy, depression, malaise, renal disease, arthritis, portal vein thrombosis, and budd chiari. The present disclosure is also directed to treating liver injury and/or reducing the likelihood of further liver injury associated with or occurring directly from or secondary to a variety of etiologies especially including hepatitis (all forms), cirrhosis (all types), non-alcoholic fatty liver diseases (NAFLD), including non-alcoholic steatohepatitis (NASH), NAFLD or NASH including primary NASH, NASH secondary to liver transplantation (NASH post-liver transplantation), preservation injury of donated organs, acute and chronic liver transplant rejection and metabolic conditions including, for example, Wilson's disease, hemochromatosis, and alpha one antitrypsin deficiency. In this method, an effective amount of an androgen receptor agonist according to the present invention is administered to a patient with liver injury and/or at risk for further liver injury as described above in order to treat, inhibit or reduce the likelihood of liver injury which occurs directly as a consequence of or secondary to one or more of the disease states and/or conditions as described above. As a consequence of the treatment methods described above, the occurrence and/or severity of one or more of the following conditions will be substantially reduced: liver failure, liver shock, obstructive jaundice, primary biliary cirrhosis, primary sclerosing cholangitis, portal hypertension, ascites, variceal bleeding, encephalopathy, depression, malaise, renal disease, arthritis, portal vein thrombosis and budd chiari.

As the skilled artisan readily recognizes the embodiments disclosed herein can be adapted to pediatric subjects e.g., subjects less than 18 years old. The doses of active ingredients used can be adjusted accordingly depending on the age and sex of the subject.

In any of the methods disclosed herein, the step of identifying a subject in need of treatment can include the step of determining the number of CAG or GGN repeats in the subject’s androgen receptor gene. The skilled artisan is capable of using this information for identifying subjects in need of treatment, providing specific doses of the androgen receptor agonists (e.g., testosterone undecanoate or testosterone tridecanoate), or both, as described herein.

Without wishing to be bound by theory, it is believed that is some aspects of the embodiments described herein, the method and compositions can normalize ratios of serum testosterone to estradiol. Thus, in some aspects, subjects are believed to have a testosterone to estradiol ratio imbalance which can be treated, and therefore become more normal.

Without wishing to be bound by theory, it is believed that is some aspects of the embodiments described herein, the methods and compositions described herein exerted surprising or expected effects in liver and related disease because they are delivered orally e.g., perorally, and absorbed via the intestinal lymphatic system. It is contemplated that delivery via the intestinal lymphatic system provides surprising or unexpected results due to one or more of the following: effects in the lymphatic or interstitial compartment, improved delivery to target tissues or cells (e.g., one or more of fat, adipocytes, preadipocytes, liver, liver cells, hepatocytes, adipose, white blood cells, red blood cells, stem cells, bone, bone cells, stem cells, androgen receptor cells, non-androgen receptor containing cells, cells involved in glucose metabolism, and cells involved in fatty acid metabolism). Thus, in some specific aspects, delivery of the compositions via the methods disclosed herein results in transport of the active agent (e.g., testosterone ester) via intestinal lymphatics, chylomicrons, chylomicron remnants to specific target tissues and cells to exert or provide the unexpected or surprising results described herein.

In some aspects of the method embodiments described herein, the treatments with oral testosterone esters in testosterone deficient subjects, raise the subject’s serum testosterone levels over baseline or e.g., into the normal or eugonadal range for from 0-2 hours, 2-4 hours, 4-6 hours, 6-8 hours, 8-10 hours, 10-12-hour, 12-14 hours, 14-18 hours, 18-20 hours, 20-22 hours, or 22-24 hours per day.

Combination Therapy

In some embodiments of the methods disclosed herein, the oral testosterone ester therapy is in conjunction with another therapeutic treatment. In one aspect, the oral testosterone therapy is administered with one or more additional therapeutic agents (not necessarily at the same time or frequency as the oral testosterone ester therapy or androgen receptor agonist therapy). In some embodiments the one or more additional therapeutic agent is a statin. In some embodiments the statin is atorvastatin, rosuvastatin, simvastatin, pravastatin, lovastatin, fluvastatin, or

pitavastatin. In one aspect, the amount of atorvastatin administered per day is from about 10 mg to about 60 mg. In one aspect, the amount of rosuvastatin administered per day is from about 5 mg to about 20 mg. In one aspect, the amount of simvastatin administered per day is from about 10 mg to about 40 mg. In one aspect, the amount of pravastatin administered per day is from about 10 mg to about 80 mg.

In one aspect, the amount of lovastatin administered per day is from about 20 mg to about 40 mg. In one aspect, the amount of fluvastatin administered per day is from about 20 mg to about 80 mg. In one aspect, the amount of pitavastatin administered per day is from about 1 mg to about 4 mg. In one aspect, the amount of atorvastatin is from about 10 mg to about 40 mg.

In one aspect, amount of rosuvastatin is from about 10 to about 20 mg. In one aspect, the amount of simvastatin is from about 10 mg to about 20 mg. In one aspect, the amount of pravastatin is from about 10 mg to about 20 mg. In one aspect, the amount of lovastatin is about 40 mg. In one aspect, the amount of fluvastatin is from about 20 mg to about 40 mg. In one aspect, the amount of pitavastatin is from about 1 mg to about 3 mg. Combination therapies can include administration of more or more of the following: a FXR agonist, a PPAR alpha ordelta agent, a lipid modulator, an anti-inflammatory (e.g., steroidal or non-steroidal anti-inflammatory agents or other), an antioxidant, an immunomodulator, an insulin senitizer, an incretin mimetic, a hemorrheologic agent, an inhibitor of apoptosis, an agonist of the peroxisome proliferator, a thyroid hormone receptor modulator, an ASK1 inhibitor, an Acetyl-CoA Carboxylase (ACC) inhibitor, a fatty-acid/bile-acid Conjugate, galectin inhibitor, caspase protease inhibitor or a combination thereof in conjunction with the androgen receptor agonist (e.g., testosterone ester like testosterone undecanoate or testosterone

tridecanoate).

The present disclosure is also directed to methods of treating hepatitis (all types, including non-alcoholic steatohepatitis (NASH)), cirrhosis (all types), fatty liver disease, including non-alcoholic fatty liver disease (NAFLD), including cirrhosis in a patient at risk, primary NASH or NASH secondary to liver transplantation, by administering an effective amount of an androgen receptor agonist compound as otherwise described hereinabove to said patient. In this aspect, a method for treating NAFLD, NASH including primary NASH, cirrhosis and/or NASH secondary to liver transplantation (NASH post-liver transplantation) comprises orally administering to a patient in need thereof an effective amount of a testosterone ester (e.g., testosterone undecanoate, testosterone tridecanoate, or a combination thereof) and vitamin E, a vitamin E prodrug or a vitamin E derivative, as otherwise disclosed herein, optionally in combination with a carrier, additive or excipient. In treating the above disease states and/or conditions there is an inhibition or a reduction in the likelihood of liver injury or that one or more of the following conditions will occur in the treated patient: liver failure, portal hypertension, ascites, variceal bleeding, encephalopathy, depression, malaise, renal disease, arthritis, portal vein thrombosis and/or budd-chiari. Thus, in one aspect, the methods described herein involve oral administration of a testosterone ester (e.g., testosterone undecanoate, testosterone tridecanoate, or a combination thereof) formulated with vitamin E, a vitamin E prodrug, or a vitamin E derivative. In a preferred aspect, the Vitamin E compound is d-alpha-tocopherol or d- alpha tocopherol acetate. In another preferred aspect, the amount of d-alpha-tocopherol (or it’s acetate) administered per day is from about 100 IU to about 2000 IU, 200 IU to about 1600 IU per day, about 400 IU to about 1000 IU per day or about 600 IU to 900 IU per day. These amounts can be co-formulated with the testosterone esters or administered separately. The exemplary formulations described herein can be adapted accordingly to use d-alpha-tocopherol (or e.g., a prodrug thereof (d-alpha-tocopheryl acetate)), tocotrienol and other related Vitamin E related compounds. Thus, in certain aspects of this embodiment a method of treating a subject in need of treatment is provided, said method comprising identifying a subject in need of treatment (e.g., has a liver disease or condition) and orally administering a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject in combination with one or more immunosuppressive agents. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating a need for combination treatment. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester improves liver disease or reduces the risk of liver disease while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject.

In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10th- 1/15th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In certain embodiments related to the treatment of liver disease, liver injury, NAFLD, NASH or cirrhosis which occurs secondary to a liver transplant, including acute and chronic transplant rejection, the compositions described hereincan be co-administered (e.g., according to the methods described herein) to the transplant patient with an effective amount at least one immunosuppressive agent chosen from Sandimmune (cyclosporine), Neoral (cyclosporine), Prograf (tacrolimus), prednisone, Imuran (azathioprine), Cellcept (mycophenolate mofetil), Zenapax (daclizumab), or Simulect (basiliximab). Thus, in certain aspects of this embodiment a method of treating a subject in need of treatment is provided, said method comprising identifying a subject in need of treatment (e.g., a subject that has had a liver transplant) and orally administering a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject in combination with one or more immunosuppressive agents. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating a need for combination treatment. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, oral administration of the testosterone ester improves liver disease or reduces the risk of liver disease while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of

testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10th- 1/15th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 45-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In other alternative embodiments, the compositions described herein can be administered to a patient where applicable (in those conditions such as NAFLD, NASH, etc. which occur as a consequence of metabolic syndrome and/or type P diabetes) in combination with an effective amount of one or more agents which are used to treat type P diabetes or metabolic syndrome including metformin, glibenclamide, gliclazide, rosiglitazone, pioglitazone, troglitazone, acarbose, miglitol, nateglinide, repaglinide, exenatide, sitagliptin, pramlintide and mixtures thereof. Thus, in certain aspects of this embodiment a method of treating a subject in need of treatment is provided, said method comprising identifying a subject in need of treatment (e.g., a subject that has liver disease or is at risk of having liver disease) and orally administering a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject in combination with an agent useful for treating type P diabetes or metabolic syndrome. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating a need for combination treatment. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect of this method, oral administration of the testosterone ester improves liver disease or reduces the risk of liver disease while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprise oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg,

750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about l/10th-l/15th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 46-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

Certain embodiments relate to the treatment of hepatitis (alcoholic and non-alcoholic), which occurs as a consequence of infections (viral and non-viral), drugs, ischemia, toxins, pregnancy, alcohol, toxins, autoimmune conditions (systemic lupus erythematosus) and metabolic conditions, including Wilson's disease, hemochromatosis and alpha one antitrypsin deficieincy. Hepatitis which may be treated according to the present invention includes hepatitis which occurs as a consequence of infectious disease, especially including a viral infection such as a hepatitis A, B, C, D or E viral infection, or hepatitis which occurs as a consequence of a cytomegalovirus, Epstein-Barr, yellow fever, mumps virus, rubella virus, herpes simplex virus, or adenovirus infection or a non-viral selection including an infection from toxoplasma, leptospira, Q fever or Rocky Mountain Spotted Fever. In this embodiment, an androgen receptor agonist (e.g., testosterone undecanoate, testosterone tridecanoate, or a combination thereof) is administered in effective amounts to a patient with a viral hepatitis infection in order to inhibit, treat or reduce the likelihood of liver injury which occurs as a consequence of that viral or non- viral infection. Compositions described herein can be administered alone or in combination with an effective amount of an anti-hepatitis infectious agent, such as an anti-viral agent, including Hepsera (adefovir dipivoxil), lamivudine, entecavir, telbivudine, tenofovir, emtricitabine, clevudine, valtoricitabine, amdoxovir, pradefovir, racivir, BAM 205, nitazoxanide, UT 231-B, Bay 41-4109, EHT899, zadaxin (thymosin alpha-1) and mixtures thereof for hepatitis B infections and MM 283, VX-950 (telaprevir), SCH 50304, TMC435, VX-500, BX-813,

SCH503034, R1626, ITMN-191 (R7227), R7128, PF-868554, TT033, CGH-759, GI 5005, MK- 7009, SIRNA-034, MK-0608, A-837093, GS 9190, ACH-1095, GSK625433, TG4040 (MVA-

HCV), A-831, F351, NS5A, NS4B, ANA598, A-689, GNI-104, IDX102, ADX184, GL59728, GL60667, PSI-7851, TLR9 Agonist, PHX1766, SP-30 and mixtures thereof for hepatitis C infections. In some embodiments, additional pharmaceutical compositions especially useful for treating hepatitis from viral infections, in particular, hepatitis b or hepatitis C infections comprise an effective amount of one or more androgen receptor agonists (e.g., testosterone undecanoate or testosterone tridecanoate) as disclosed herein in combination with at least one agent selected from the group consisting of hepsera (adefovir dipivoxil), lamivudine, entecavir, telbivudine, tenofovir, emtricitabine, clevudine, valtoricitabine, amdoxovir, pradefovir, racivir, BAM 205, nitazoxanide, UT 231-B, Bay 41-4109, EHT899, zadaxin (thymosin alpha-1) and mixtures thereof for hepatitis B infections and NM 283, VX-950 (telaprevir), SCH 50304, TMC435, VX- 500, BX-813, SCH503034 (boceprevir), R1626, ITMN-191 (R7227), R7128, PF-868554,

TT033, CGH-759, GI 5005, MK-7009, SIRNA-034, MK-0608, A-837093, GS 9190, ACH-1095, GSK625433, TG4040 (MVA-HCV), A-831, F351, NS5A, NS4B, ANA598, A-689, GNI-104, IDX102, ADX184, GL59728, GL60667, PSI-7851, TLR9 Agonist, PHX1766, SP-30 and mixtures thereof, in combination with a pharmaceutically acceptable carrier, additive or excipient.

Biomarkers

Any appropriate biomarker can be used in the methods and compositions described herein (including imaging analysis, histology, genetic, microRNA and the such). Specific biomarkers are described herein in the method embodiments and Examples. Additional biomarkers, including some that are described herein elsewhere, are illustrated below.

In one embodiment, the methods and compositions described herein, are useful for treating a subject in need of treatment e.g., having NASH or suspected of having NASH or having fatty liver disease or having liver disease. Thus, the methods and compositions described herein can be used to treat NASH (or suspected of having NASH or having fatty liver disease or having liver disease) of any grade or type. In one aspect, the methods and compositions as disclosed herein, reduce liver fat (or e.g., steatosis) or reduce the rate of increase in liver fat (or steatosis). In one aspect, the methods and compositions can reduce liver fat by greater than 5%, 10%, 15%, 20%, 25% or 30% in at least 1, 5, 10, 20, 30, 40, or 50% of subjects treated. In one aspect, the biomarker (liver fat) is determined as a % mean relative reduction in liver fat reduction with MRI-PDFF (Magnetic Resonance Imaging-Derived Proton Density Fat Fraction). In one aspect, the methods and compositions reduce liver triglyceride concentration or decrease the rate of increase liver triglyceride concentration. In one aspect, the biomarker (liver triglyceride concentration) is measured by NMRS (magnetic resonance spectroscopy). In one aspect, the method and composition can improve or ameliorate diffuse liver disease due to liver fat or iron deposition by 5%, 10%, 15%, 20% or 25% or more. In one aspect, non-invasive magnetic resonance imaging (MRI-PDFF) is sued for the evaluation of liver fat or iron deposition. In one aspect, the methods and compositions disclosed herein, ameliorates fatty infiltration (e.g., reduces fatty infiltration or reduces the rate of increase of fatty infiltration). In one aspect, abdominal ultrasound is used to determine fatty infiltration. In one aspect, CT (computed tomography) is used to assess the liver biomarker. In one aspect, the methods and compositions described herein improve liver stiffness (LS)/fibrosis. In one aspect, improved liver stiffness/fibrosis is at least a 5, 10, 15, 20 or 25% reduction in MRE (Magnetic Resonance Elastography)-stiffness. In one aspect, ultrasound based elastography is used for the assessment of liver tissue stiffness in fibrosis. In one aspect, transient elastography (TE) (e.g., Fibroscan®, Echosens, Paris) is used (ultrasound-based elastography) to determine liver stiffness. In one aspect, pSWE (Elastography point quantification, ElastPQ™, Phillips) or ARFI imaging (Virtual touch tissue quantification™, Siemens) is (ultrasound-based elastography techniques) used to assess liver stiffness. In one aspect, liver stiffness is monitored via Real Time TE (RTE) which is a qualitative assessment of liver stiffness. Magnetic resonance elastography (MRE) can determine liver stiffness by analysis of mechanical waves propagating through the liver. In one aspect, the methods and compositions disclosed herein reduce liver stiffness by 5, 10, 20, 30, 40, 50, 60 or 75% or more. In one aspect, the methods and compositions disclosed herein reduce the rate of increase of liver stiffness by 5, 10, 20, 30, 40, 50, 60 or 75% or more. In one aspect, the method and compositions disclosed herein improve on liver fibrosis as measured by non-invasive LiverMultiscan. In one aspect, the method and compositions reduce hepatic steatosis as monitored by imaging studies along with decreasing in ALT, CK18, or both. In one aspect, the methods and compositions described herein improve or ameliorate liver fibrosis as assed by histology and reduces risk of progression to cirrhosis. In one aspect, the methods and compositions described herein improve by at least 1 or 2 points (e.g., decrease) the NAFLD activity score (NAS). In one aspect, the methods and compositions described herein improve at least a point reduction in either lobular inflammation or hepatocellular ballooning. In one aspect, the methods and compositions improve or resolve NASH on overall histopathological interpretation by an experienced pathologist (e.g., target score of 0 on ballooning and 0 or 1 for inflammation, but no greater than 1) and no worsening of fibrosis (e.g., no one stage increase in on fibrosis score). In one aspect, the methods and compositions described herein reverse NASH (e.g., change from NAS score from 5 to 3) with no evidence of progression to advanced fibrosis

(e.g., stage 3 or 4). In one aspect, the methods and compositions described herein improve fibrosis (e.g., lessen) without worsening of NAS or no progression of steatohepatitis (or reduce progression of steatohepatitis). In one aspect, the methods and compositions described herein do not increase NAFLD activity as assessed by NAS and lessen or reverse fibrosis. In one aspect, the methods and compositions described herein when used in a population of patients, at least 10%, 20%, 30% 40% or 50% or more achieve a > 1 -Stage Improvement in Fibrosis According to the NASH Clinical Research Network (CRN) Classification Without Worsening of NASH. In one aspect, the methods and compositions described herein prevent the development of cirrhosis or lessen the likelihood of progression to cirrhosis. In one aspect, the methods and compositions described herein slowing histological progression to cirrhosis. In one aspect, the methods and compositions described herein improve event free survival (e.g., as assessed by all-cause mortality, new decompensation events, MELD score progression). In one aspect, the methods and compositions described herein decrease in all-cause mortality. In one aspect, the methods and compositions described herein decrease in liver specific mortality. In one aspect, the methods and compositions described herein decrease liver transplantation rates. In one aspect, the methods and compositions described herein increase 12 month survival rates. In one aspect, the methods and compositions described herein increase event free survival (EFS) at week 52 as Assessed by Time to the First Clinical Event. In one aspect, the methods and compositions described herein decrease events/rate of events of decompensation in compensated patients. In one aspect, the methods and compositions described herein decrease rate of ascites events or ascites grade. In one aspect, the methods and compositions described herein decrease rate of HE or progress in HE (e.g., as assessed by MRI). In one aspect, the methods and compositions described herein decrease in hospital admissions or hospital admission rates. In one aspect, the methods and compositions described herein decrease unscheduled clinic or ER visit. In one aspect, the methods and compositions described herein decrease the number of tests performed. In one aspect, the methods and compositions described herein decrease lost work days. In one aspect, the methods and compositions described herein decrease infection rates or number of events. In one aspect, the methods and compositions described herein improve the Child-Pugh- Turcotte Score. In one aspect, the methods and compositions described herein lower Progression from A to B In one aspect, the methods and compositions described herein do not worsen CPT. In one aspect, the methods and compositions described herein improve CPT score by at least 1 or 2 points. In one aspect, the methods and compositions described herein improve MELD score.

In one aspect, the methods and compositions described herein lessen responder progression to higher MELD score. In one aspect, the methods and compositions described herein do not worsen MELD score. In one aspect, the methods and compositions described herein improve MELD score by at least one or two points. In one aspect, the methods and compositions described herein lower % of subjects achieving transplantation qualifying MELD score of 14. In one aspect, the methods and compositions described herein increase in MELD score from <12 to 15 or higher. In one aspect, the methods and compositions described herein improve the hepatic venous pressure gradient (HVPG). In one aspect, the methods and compositions described herein reduce of proportion of subjects that progress to HVPG >10 mm. In one aspect, the methods and compositions described herein lower HVPG <10mm. In one aspect, the methods and compositions described herein reduce HVPG by 5, 10, 15, 20, 25 or 30% or more. In one aspect, the methods and compositions described herein improve body composition. In one aspect, the methods and compositions described herein improve bone density (e.g., one or more of femoral, lumbar, neck, and total bone mass. In one aspect, the methods and compositions described herein improve anemia. In one aspect, the methods and compositions described herein improve gynecomastia (e.g., lessen). In one embodiment, a method of reducing progression or the risk of progression of NASH to cirrhosis a subject having or suspected of having NASH is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment e.g., has NASH or is at risk of having NASH progressing to cirrhosis. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating an increased risk of NASH progressing to cirrhosis. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect of this method, oral administration of the testosterone ester reduces the progression of NASH to cirrhosis while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg,

750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about l/10th-l/15th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 46-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities:

obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia. In one aspect, the methods and compositions disclosed herein improve or ameliorate, changes in body composition, muscle mass, appendicular lean mass, total lean mass, fat mass, high VAT (visceral adipose fat), waist circumference, weight gain, change in BMI,

mobility/frailty: (e.g., timed up and go-TUG (mobility) score, hand grip strength, liver frailty index as measured by score in functional assessments of grip strength (kg), balance (seconds), and chair stands (seconds)), PROs: functional status of patient-COA-symptoms only known to patients, QOL (quality of life) assessed by the CLDQ, perceived HRQoL score, depression, mobility, sexual dysfunction, and FIS fatigue questionnaire, in a subject in need of treatment (e.g., as described in the method embodiments described herein and specifically subjects having cirrhosis, NASH, NAFLD, steatohepatitis, liver disease). In one embodiment, a method of reducing progression or the risk of progression of NASH to cirrhosis a subject having or suspected of having NASH is provided said method comprising oral administration of a pharmaceutical composition having a testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) to said subject. In one aspect, the subject is in need of treatment e.g., has NASH or is at risk of having NASH progressing to cirrhosis. In one aspect, the subject in need of treatment has one or more biomarkers (or clinical characteristics) indicating an increased risk of NASH progressing to cirrhosis. In one aspect, the one or more biomarkers are outside the normal range. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect, said subject in need of treatment has testosterone deficiency with serum testosterone levels of less than 350 ng/dL, 300 ng/dL, 250 ng/dL, 200 ng/dL, 150 ng/dL or 100 ng/dL. In one aspect, the one or more biomarkers are above the normal range but between 2-3 times the upper limit of the normal range or more. In one aspect, the one or more biomarkers are above the normal range but below 2 times the upper limit of the normal range. In one aspect of this method, the subject has testosterone deficiency. In one aspect of this method, oral administration of the testosterone ester reduces the progression of NASH to cirrhosis while not substantially increasing serum levels of or decreases serum levels of lipoprotein-associated phospholipase A2. In one aspect, said method increases said subject’s serum testosterone levels over their baseline. In another aspect said method increases said subject’s serum testosterone levels into the normal range. In one aspect, said method comprises oral administration of 200 mg to 750 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 300 mg to 600 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of 450 mg of testosterone undecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 400 mg to 2000 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 600 mg to 1800 mg of testosterone tridecanoate per day to a male subject. In one aspect, said method comprises oral administration of about 500 mg, 750 mg, 1000 mg, or 1250 mg of testosterone tridecanoate per day to a male subject. These dose ranges are typically those for male subjects whereas the female dose range corresponds to about 1/10th- 1/15th of these values. In one aspect the subject is from 18-25 years old, 26-35 years old, 36-45 years old, 46-55 years old, 56-65 years old or older than 65 years old. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, triglycerides, LDL, cholesterol, liver biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, any specific biomarkers disclosed in the Tables of Example 1, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers. In one aspect of this embodiment, the subject has one or more of the following comorbidities: obesity, type 2 diabetes, dyslipidemia, cardiovascular disease, thyroid dysfunction, chronic kidney disease, liver disease, osteoporosis, hypogonadism, hypertension, sarcopenia, or cachexia.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more serum biomarkers.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more lipogenesis/ lipid transporters biomarkers.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more lipogenesis/ lipid transporters biomarkers chosen from acetyl-coA carboxylase (ACC), fatty acid transport protein-5 (FATP-5) or the fatty acid transporter CD36, liver-fatty acid binding protein (FABP), Ceramides, 3-nitrotyrosine (oxidative stress marker), HNE (4-hydroxy-2-noneal, a marker of lipid peroxidation), 8-hydroxydeoxyguanosine (a marker of oxidative DNA damage), Glutamate dehydrogenase (mitochondrial marker enzyme) , Glucose 6-phosphatase (microsomal enzyme marker), LD or LDH (Lactate dehydrogenase), and resitsin. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more lipo-Apoptosis biomarkers e.g., CK-18 (cytokeratin 18) Cathepsin B, caspase cleaved CK18, flCK, caspase 3/7, platelet count, full blood cell count ( e.g., thrombocytopenia, al -antitrypsin, ferritin, and adipokines (e.g., adiponectin and leptin).

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more metalloproteinase biomarkers e.g., (MMPs)-MMPs 2, MMP-3, and MMP-9.

In one aspect, the methods and compositions described herein are utilized in conjunction with transforming growth factor (TOR)-bI as a biomarker.

In one aspect, the methods and compositions described herein are utilized in conjunction with one or more tissue inhibitors of metalloproteinases (TIMPs) as biomarkers e.g., ΊΊMR- l,TIMP-2, Microfibril-associated protein 4 (MFAP4).

In one aspect, the methods and compositions described herein are utilized in conjunction with hyaluronic acid (HA) N Glycans profiles as biomarkers.

In one aspect, the methods and compositions described herein are utilized in conjunction with one or more of the following biomarkers: Laminin, Procollagen type IP amino-terminal peptide (RIPNR), Chitinase-3-like protein 1 (CHDL1 or YKL-40), CTGF (connective tissue growth factor), and Collagen type IV-S or VI.

In one aspect, the methods and compositions described herein are utilized in conjunction with pro-inflammation and pro-fibrotic mediators as biomarkers.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more of hs-CRP or cytokine biomarkers. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more biomarkers chosen from IL-1,IL-1 beta, NF-KappaB, MCP-l(CCL2-Chemokine) IL-6, IL-8, RBP-4, soluble CD14, TNF-alpha, MCP-1, leptin, visfatin, adiponectin, fibrinogen,

fibrinonectin, collagens (I-IP), undulin, elastin, proteoglycans, PICP (procollagen type 1 carboxy terminal peptide), ICAM, VCAM (adhesion molecules), and INK (protein kinase). In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more biomarkers that are liver enzymes. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more of alanine amino transferase (ALT), aspartate amino transferase (AST), alkaline phosphatase (ALP), and gamma glutamyl transferase (GGT).

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more biomarkers of synthetic function. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more biomarkers of synthetic function chosen from prothrombin time (PT/INR), bilirubin,

haptoglobin, hemoglobin, albumin, Apolipoprotein Al, o2-macroglobulin, ceruloplasmin, transferrin and hepcidin, sodium serum alpha protein, circulating ammonia, and creatinine.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more hormone or endocrine biomarkers. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more hormone or endocrine biomarkers chosen from Prolactin, LH, FSH, E, TT, freeT, DHT, SHBG, oestrone, thyroid panel, and cortisol.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more hypertension related biomarkers.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more diabetes related biomarkers. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more diabetes related biomarkers chosen from HB1AC, Impaired Fasting glucose, fasting insulin, and HOMA- IR (Homeostasis Model Assessment-insulin resistance).

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more anemia related biomarkers. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more anemia related biomarkers chosen from hemoglobin levels, hematocrit levels, complete blood count (CBC), and MCV (e.g., a measure of the average size of red blood cells). In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more dyslipidemia related biomarkers. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more dyslipidemia related biomarkers chosen from HDL, LDL, total cholesterol, triglycerides, non HDL cholesterol, VLDL, saturated free fatty acid, unsaturated free fatty acid, total free fatty acid, ApoB, and apolipoprotein Al.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more sarcopenia related biomarkers. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more sarcopenia related biomarkers chosen from decrease in fat mass relative to fat free mass

(includes muscle mass), DEXA scan for bone mineral, fat, bone-mineral-fat-free mass, fat-free skeletal muscle mass e.g., obtained with an imaging technique (such as computerized tomography (CT) and magnetic resonance imaging (MRI)), bioimpedance analysis (BIA) for muscle mass.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three of creatinine, 3-methylhistidine, and urinary creatinine excretion.

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more metabolic syndrome related biomarkers e.g., sBP >130, hyper glycemia-fasting glucose >100, waist circumference >40 in men, TG >150, HDL <40

In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more combination biomarkers of liver fibrosis. In one aspect, the methods and compositions described herein are utilized in conjunction with one, two or three or more combination biomarkers of liver fibrosis chosen from AST/ALT ratio, BARD related (e.g., BMI, AST, ALT, and Diabetes Mellitus), and e.g., combinations of FIB-4, Platelet count, AST, ALT, and age.

Other biomarkers for use in the methods include, but are not limited to:

Fibrometer test- Platelet count, prothrombin index, AST, o2-macro-globulin, hyaluronic acid, urea, age

Fibrometer A- Prothrombin index,' o2 macroglobulin, hyaluronic acid, age Fibrotest (FT)- Haptoglobin, o2-macro-globulin, apolipoprotein Al. GGT, bilirubin, age, gender

NAFLD Fibrosis Score (NFS)- Age, BMI, platelets, albumin, AST/ALT, IFG /diabetes

Fibrospect-P- Hyaluronic acid, TIMP-1, o2-macroglobulin PGA-index- Prothrombin time, GGT, apolipoprotein Al

PGAA-index- Prothrombin time, GGT, apolipoprotein Al, o2-macroglobulin

Pohl score- AST/ALT-ratio, platelet coimt

ELF score- Hyaluronic acid, TIMP-1, age, MMP-3

SHASTA- HA, AST, albumin Fibrosis probability-index, FPI- Age, AST, cholesterol, insulin resistance (HOMA), past alcohol intake

APRI score- AST, platelet coimt alpha2-macroglobulin, age, gamma glutamyl transpeptidase, and hyaluronic acid.

Other Methods

A method of diagnosing NAFLD in a subject is provided herein said method comprising: determining the subjects serum testosterone level, determining the subjects BMI, or both, wherein a subject having low testosterones levels and a BMI greater than or equal to 30, or the subject is obese, is diagnosed with NAFLD. According to this method, a subject diagnosed with NAFLD is furthered assessed by analyzing one or more biomarkers or performing imaging studies to confirm diagnosis and stage as well as determine treatment. For example, a subject diagnosed with liver is further assessed by MRI-PDFF or MRE, or one or more serum or NAS biomarkers are assessed. In one aspect, the biomarker is one or more chosen from ALP, ALT, AST, GGT, TRIGLYCERIDES, LDL, Cholesterol, Liver Biopsy, inflammation biomarkers, non-HDL cholesterol, hematocrit, hemoglobin, lipoprotein phospholipase A2, bilirubin, albumin, SHBG, imaging biomarkers, liver histology biomarkers, biomarkers in the literature related to the disease and condition described herein, and liver damage biomarkers.

In one embodiment, the subject diagnosed with the method described herein (in those conditions such as NAFLD, NASH, etc. or which occur as a consequence of metabolic syndrome and/or type P diabetes) can be treated for NAFLD or NASH with of one or more agents which are used to treat type P diabetes or metabolic syndrome including metformin, glibenclamide, gliclazide, rosiglitazone, pioglitazone, troglitazone, acarbose, miglitol, nateglinide, repaglinide, exenatide, sitagliptin, pramlintide and mixtures thereof.

In one embodiment, the subject diagnosed (e.g., hypogonadal, obese, having fatty liver, having live fat greater than 8%, MRE, NAFLD, NASH, one or more serum biomarkers described herein, etc.) with the method described herein can be treated for NAFLD or NASH with of one or more pharmaceutical agents that inhibits c-Jun N-terminal kinase, p38 mitogen-activated protein kinase, or both.

In one embodiment, the subject diagnosed (e.g., hypogonadal, obese, having fatty liver, having live fat greater than 8%, MRE, NAFLD, NASH, one or more serum biomarkers described herein, etc.) with the method described herein can be treated for NAFLD or NASH with of one or more pharmaceutical agents that inhibits ASK1 kinase. One example of such an agent is selonsertib.

In one embodiment, the subject diagnosed (e.g., hypogonadal, obese, having fatty liver, having live fat greater than 8%, MRE, NAFLD, NASH, one or more serum biomarkers described herein, etc.) with the method described herein can be treated for NAFLD or NASH with of one or more pharmaceutical agents is an agonist of famesoid X receptor. One example of such an agent is obeticholic acid.

In one embodiment, the subject diagnosed (e.g., hypogonadal, obese, having fatty liver, having live fat greater than 8%, MRE, NAFLD, NASH, one or more serum biomarkers described herein, etc.) with the method described herein can be treated for NAFLD or NASH with of one or more pharmaceutical agents is an inhibitor of the peroxisome proliferator activated receptor alpha, delta, or both. One example of such an agent is elafibrinor. In one embodiment, the subject diagnosed (e.g., hypogonadal, obese, having fatty liver, having live fat greater than 8%, MRE, NAFLD, NASH, one or more serum biomarkers described herein, etc.) with the method described herein can be treated for NAFLD or NASH with of one or more pharmaceutical agents is an antagonist of C-C chemokine receptor types 2, 5, or both. One example of such an agent is cenicriviroc.

In one embodiment, the subject diagnosed (e.g., hypogonadal, obese, having fatty liver, having live fat greater than 8%, MRE, NAFLD, NASH, one or more serum biomarkers described herein, etc.) with the method described herein can be treated for NAFLD or NASH with of one or more pharmaceutical agents that is chosen from butanoic acid, CER209, evogliptin, DUR928, MK-4074, OPRX-106, PF06865571, PF06882961, PXS-5382A, RG-125, RYI-018, seladelpar, SGM-1019, TVB-2640, or a combination thereof.

In one embodiment, the subject diagnosed (e.g., hypogonadal, obese, having fatty liver, having live fat greater than 8%, MRE, NAFLD, NASH, one or more serum biomarkers described herein, etc.) with the method described herein can be treated for NAFLD or NASH with of one or more pharmaceutical agents that is chosen from aramchol, ARX618, BI 1467335, DS102, EDP-305, Emricasan, gemcabene, GR-MD-02, GRI-0621, GS-0976, GS-9674, IMM-124E, IONIS-DGAT2Rx, IVA-337, Lipaglyn, LJN452, LMB763, MGL-3196, MN-001, MSDC- 0602K, NC101, NGM282, NS-0200, Ozempic, PF-05221304, PF-06835919, remogliflozin etabonate, SHP626, TVB-2640, VK2809, or a combination thereof.

In one embodiment, the subject diagnosed (e.g., hypogonadal, obese, having fatty liver, having live fat greater than 8%, MRE, NAFLD, NASH, one or more serum biomarkers described herein, etc.) with the method described herein can be treated for NAFLD or NASH with of one or more pharmaceutical agents that is chosen from cenicriviroc, elafibranor, ocaliva (obeticholic acid), selonsertib, or a combination thereof.

In one embodiment, the subject diagnosed (e.g., hypogonadal, obese, having fatty liver, having live fat greater than 8%, MRE, NAFLD, NASH, one or more serum biomarkers described herein, etc.) with the method described herein can be treated for NAFLD or NASH with of one or more pharmaceutical agents is an androgen receptor agonist. One example of such an agent is testosterone or a testosterone ester (and in a preferred aspect testosterone or testosterone ester formulated for and delivered orally).

In some embodiments of the methods disclosed herein, the subject diagnosed according to the instant disclosure is administered oral testosterone ester therapy. In one aspect, the oral testosterone therapy is administered with one or more additional therapeutic agents (not necessarily at the same time or frequency as the oral testosterone ester therapy or androgen receptor agonist therapy).

In some embodiments the one or more additional therapeutic agent is a statin. In some embodiments the statin is atorvastatin, rosuvastatin, simvastatin, pravastatin, lovastatin, fluvastatin, or pitavastatin. In one aspect, the amount of atorvastatin administered per day is from about 10 mg to about 60 mg. In one aspect, the amount of rosuvastatin administered per day is from about 5 mg to about 20 mg. In one aspect, the amount of simvastatin administered per day is from about 10 mg to about 40 mg. In one aspect, the amount of pravastatin

administered per day is from about 10 mg to about 80 mg. Combination of the other

pharmaceutical agents described herein with a statin and another agent instead of the testosterone or testosterone ester are also contemplated.

In one aspect, the amount of lovastatin administered per day is from about 20 mg to about 40 mg. In one aspect, the amount of fluvastatin administered per day is from about 20 mg to about 80 mg. In one aspect, the amount of pitavastatin administered per day is from about 1 mg to about 4 mg. In one aspect, the amount of atorvastatin is from about 10 mg to about 40 mg.

In one aspect, amount of rosuvastatin is from about 10 to about 20 mg. In one aspect, the amount of simvastatin is from about 10 mg to about 20 mg. In one aspect, the amount of pravastatin is from about 10 mg to about 20 mg. In one aspect, the amount of lovastatin is about 40 mg. In one aspect, the amount of fluvastatin is from about 20 mg to about 40 mg. In one aspect, the amount of pitavastatin is from about 1 mg to about 3 mg.

Combination therapies (or monotherapies) can include administration of more or more of the following with an androgen receptor agonist or modulator as described herein: a FXR agonist, a PPAR alpha or delta agent, a lipid modulator, an anti-inflammatory (e.g., steroidal or non-steroidal anti-inflammatory agents or other), an antioxidant, an immunomodulator, an insulin senitizer, an incretin mimetic, a hemorrheologic agent, an inhibitor of apoptosis, an agonist of the peroxisome proliferator, a thyroid hormone receptor modulator, an ASK1 inhibitor, an Acetyl-CoA Carboxylase (ACC) inhibitor, a fatty-acid/bile-acid Conjugate, galectin inhibitor, caspase protease inhibitor or a combination thereof in conjunction with the androgen receptor agonist (e.g., testosterone ester like testosterone undecanoate or testosterone

tridecanoate). Combination therapies also include e.g., an androgen, testosterone, testosterone ester, testosterone undecanoate, testosterone tridecanoate with a glp-l agonist or modulator (e.g., liraglutide or semaglutide) or slgt-2 inhibitors (e.g., gliflozins, canagliflozin, ertugliflozin, dapagliflozin, empagliflozin, and sotagliflozin). Subjects for Further Diagnostic/Prognostic Testing and Treatment

As described herein methods are provided for diagnosis/prognosis and treatment of liver disease, particularly fatty liver disease, NAFLD, NASH, Cirrhosis, and symptoms thereof. The treatments and diagnosis/prognosis can be performed for subject or samples obtained from subjects as described in the below paragraph or as deemed appropriate by a medical professional. For example, the subject, as identified below, is assessed for liver fat imaging (e.g., by MRI- PDFF) or liver stiffness (MRE) or a serum sample is assessed or for one or more biomarkers described herein. This information is then used to further diagnose the subject and also can be used determine the most effective treatment.

In one aspect, the subject is a male hypogonadal subject. In one aspect, the subject is a male hypogonadal subject with class I, P, or m obesity. In one aspect, the subject is a male hypogonadal subject with class I, P, or IP obesity. In one aspect, the subject is a male hypogonadal subject having elevated or above-normal serum triglycerides. In one aspect, the subject is a male hypogonadal subject having elevated or above normal serum AST or ALT levels. In one aspect, the subject is a male hypogonadal subject having class I, P, or IP obesity and elevated or above normal serum triglycerides. In one aspect, the subject is a male hypogonadal subject having class I, P, or IP obesity and elevated or above normal serum ALT or AST. In one aspect, the subject is a male hypogonadal subject having class I, P, or IP obesity and elevated or above normal serum triglycerides, and elevated or above normal serum ALT or AST. Pharmaceutical Compositions

In certain embodiments, provided herein is a pharmaceutical composition comprising at least one steroidal compound (e.g., testosterone, dihydrotestosterone, estradiol, or analogs or prodrugs thereof) and at least one pharmaceutically acceptable carrier. In specific embodiments, the steroidal compound is a steroidal androgen (e.g., testosterone, dihydrotestosterone, or prodrugs thereof). In some embodiments, the steroidal compound is an alkylated, hydroxy- alkylated and/or hydroxy-alkoylated natural steroid (e.g., testosterone alkyl ester,

dihydrotestosterone alkyl ester, estradiol alkyl ester, or the like). In certain embodiments, analogs or prodrugs of testosterone include, e.g., esters of testosterone. In specific embodiments, the esters of testosterone include, e.g., alkyl (e.g., straight chain, branched, cyclic, unsaturated, partially saturated, fully saturated and the like) esters of testosterone. Specifically, alkyl esters of testosterone include, by way of non-limiting example, lower alkyl esters (e.g., testosterone C2- C13 alkyl esters such as testosterone propionate, testosterone enthanate, or testosterone undecanoate), or higher alkyl esters (e.g., testosterone C14+ alkyl esters such as testosterone palmitate). In further embodiments, the alkyl esters of testosterone include, by way of nonlimiting example, cycloalkylalkyl esters (e.g., testosterone cypionate), cycloalkyl esters, and alkylcycloalkyl esters. In more specific embodiments, the testosterone alkyl ester is testosterone undecanoate. In some embodiments, the at least one steroidal compound comprises (1) a testosterone lower alkyl ester (e.g., testosterone propionate, testosterone enanthate, or testosterone undecanoate); and (2) a testosterone higher alkyl ester (e.g., testosterone palmitate). Generally, as used herein, a pharmaceutical composition comprising a steroidal compound includes the disclosure of a pharmaceutical composition comprising one or more steroidal compounds.

In some embodiments, dihydotestosterone or a dihydrotestosterone ester can be used in place or in conjunction with a testosterone ester as described herein. Exemplary

dihydrotestosterone esters include, but are not limited to, dihydrotestosterone propionate, dihydrotestosterone enanthate, dihydrotestosterone cypionate, dihydrotestosterone undecanoate, dihydrotestosterone decanoate, dihydrotestosterone dodecanoate, dihydrotestosterone

tridecanoate, dihydrotestosterone tetradecanoate, and dihydrotestosterone palmitate. In certain embodiments, any pharmaceutical composition described herein comprises a therapeutically effective amount of at least one steroidal compound (e.g., a testosterone alkyl ester, such as testosterone undecanoate). In some embodiments, a therapeutically effective amount of a steroidal compound (e.g., a testosterone alkyl ester, such as testosterone

undecanoate) is divided into one or more oral dosage form. In some embodiments, the one or more of the oral dosage forms described herein collectively comprise a therapeutically effective amount of a testosterone alkyl ester (e.g., testosterone undecanoate). Thus, in some

embodiments, the therapeutically effective amount of a steroidal compound (e.g., a testosterone alkyl ester, such as testosterone undecanoate) within a pharmaceutical composition described herein may vary when the pharmaceutical composition is administered in combination with another therapy. Furthermore, therapeutically effective amounts of a formulation may depend on the specific formulation within which the at least one steroidal compound is found. For example, in some embodiments, more than one steroidal compound is present in a pharmaceutical composition described herein. Thus, when there is a combination of steroidal compounds, in certain instances one or both of the steroidal compounds present has a therapeutically effective amount that is lower than is required when the steroidal compounds are administered separately or alone. In some embodiments, a pharmaceutical composition described herein further comprises an adjuvant, which, in certain instances, allows for a lower amount of a steroidal compound to be utilized as a therapeutically effective amount.

As described in typical total daily dose ranges for adult male and female subjects are given for testosterone undecanoate and testosterone tridecanoate. Based on the doses, doses for other testosterone esters can be estimated on the T-equivalent dose which is determined e.g., by the amount of testosterone per testosterone undecanoate molecule. For example, for calculation purposes, 1 mg of T is equivalent to: 1.39 mg T-enanthate; 1.58 mg T-undecanoate; 1.43 mg T- cypionate ab so on. Although the conversion is not exact due to various properties, a skilled artisan understand how to estimate a dose of one testosterone ester for another testosterone ester based on this calculation.

In certain embodiments, a pharmaceutical composition described herein comprises about 1 mg to about 1.5 g, about 10 mg to about 1000 mg, or about 10 mg to about 200 mg of a steroidal compound (e.g., a testosterone alkyl ester, such as testosterone undecanoate). In specific embodiments, a pharmaceutical composition described herein comprises about 10 mg to about 50 mg, about 15 mg to about 40 mg, about 20 mg, to about 30 mg, or about 25 mg of steroidal compound (e.g., a testosterone alkyl ester, such as testosterone undecanoate). In other embodiments, a pharmaceutical composition described herein comprises about 70 mg to about 150 mg, about 80 mg to about 140 mg, about 90 mg to about 140 mg, about 100 mg to about 130 mg, about 110 mg to about 130 mg, about 110 mg to about 120 mg, about 130 mg to about 180 mg, about 180 mg to about 230 mg, about 230 mg to about 280 mg, about 280 mg to about 330 mg, about 330 mg to about 380 mg, about 380 mg to about 430 mg, about 430 mg to about 480 mg, about 480 mg to about 530 mg , or about 530 mg to about 580 mg of a steroidal compound (e.g., a testosterone alkyl ester, such as testosterone undecanoate). In some embodiments, a pharmaceutical composition described herein comprises about 0.1 mg to about 10 mg of a steroidal compound (e.g., a testosterone alkyl ester such as testosterone undecanoate) per kg of an individual to whom the oral dosage form is to be administered. In certain embodiments, a pharmaceutical composition described herein comprises an amount of a steroidal compound (e.g., a testosterone alkyl ester, such as testosterone undecanoate) sufficient to provide about 1 mg to about 1 g, about 5 mg to about 500 mg, about 10 mg to about 300 mg, or about 20 to about 250 mg of a steroidal compound (e.g., a testosterone alkyl ester, such as testosterone

undecanoate) to an individual upon once a day, twice a day, three times a day, or four times a day oral administration.

In some embodiments, the at least one pharmaceutically acceptable carrier is any carrier suitable for delivering an efficacious amount of a steroidal compound, e.g., a testosterone alkyl ester, to an individual. In some embodiments, the at least one pharmaceutically acceptable carrier is or comprises a hydrophilic carrier (e.g., a hydrophilic surfactant or hydrophilic additive). In certain embodiments, the at least one pharmaceutically acceptable carrier is a lipophilic carrier (e.g., a lipophilic surfactant or lipophilic additive). In some embodiments, the at least one pharmaceutically acceptable carrier is a hydrophilic carrier (e.g., a hydrophilic surfactant or hydrophilic additive) and a lipophilic carrier (e.g., a lipophilic surfactant or lipophilic additive). In certain embodiments, the hydrophilic carrier is a hydrophilic triglyceride. In specific embodiments, the hydrophilic triglyceride is a polyoxylated castor oil, or a polyoxylated hydrogenated castor oil. In some embodiments, any pharmaceutical composition provided herein consists essentially of a lipophilic carrier or combination of lipophilic carriers. In certain embodiments, any pharmaceutical composition provided herein comprises a lipophilic carrier and less than 10% w/w, less than 5% w/w or is substantially free of a hydrophilic carrier. In certain embodiments, any pharmaceutical composition provided herein comprises a lipophilic carrier and less than 10% w/w, less than 5% w/w or is substantially free of a hydrophilic carrier. In some embodiments, the pharmaceutical composition comprising a carrier (e.g., a hydrophilic carrier and/or a lipophilic carrier), the pharmaceutical composition is a solid, a semi-solid, a gel, a jelly, a paste, or the like. In certain embodiments, e.g., wherein a pharmaceutical composition comprising a hydrophilic carrier and/or a lipophilic carrier, a viscosity enhancing agent or a solidifying agent is utilized to afford a pharmaceutical composition that is a solid, a semi-solid, a gel, a jelly, a paste, or the like. Thus, in certain embodiments, the at least one pharmaceutically acceptable carrier is a hydrophilic carrier (e.g., a hydrophilic surfactant or hydrophilic additive) and a viscosity enhancing or solidifying agent. In certain embodiments, the at least one pharmaceutically acceptable carrier is a lipophilic carrier (e.g., a lipophilic surfactant or lipophilic additive) and a viscosity enhancing or solidifying agent. In some embodiments, the at least one pharmaceutically acceptable carrier is or comprises a hydrophilic carrier (e.g., a hydrophilic surfactant or hydrophilic additive), a lipophilic carrier (e.g., a lipophilic surfactant or lipophilic additive), and a viscosity enhancing or solidifying agent. In some embodiments, the at least one pharmaceutically acceptable carrier is or comprises an amphiphilic or zwitterionic carrier (e.g., a ampiphilic surfactant or ampiphilic additive). In certain embodiments, the pharmaceutically acceptable carrier is any carrier suitable for achieving one or more of the pharmacokinetic and/or pharmacodynamic profiles set forth herein.

Additives useful herein include chemical substances that are generally pharmacologically inactive. Further, the additive may be solid, liquid or semi-solid in nature at about ambient room temperature. Furthermore, the additive may be hydrophilic or lipophilic. In certain instances, a “hydrophilic additive” is a substance that has at least one polar side group in its chemical structure which will attract water; whereas a“lipophilic additive” exhibits a tendency to repel water.

In some embodiments, the hydrophilic or lipophilic additive is contained within the components forming a composition and/or pharmaceutical dosage form thereof. In certain embodiments, the hydrophilic or lipophilic additive is in an encapsulation coat in compositions. Alternatively, the additives can be comprised in the pharmaceutical composition but not as part of the composition itself. Specific, non-limiting examples of additives are described below.

Suitable additives include any additive that can facilitate the processes involving the preparation of a pharmaceutical composition and/or dosage form described herein. In some instances, such additives include those commonly utilized to facilitate the processes involving the preparation of a composition and/or a pharmaceutical dosage form described herein. These processes include agglomeration, air suspension chilling, air suspension drying, balling, coacervation, comminution, compression, pelletization, cryopelletization, encapsulation, extrusion, granulation, homogenization, inclusion complexation, lyophilization,

nanoencapsulation, melting, mixing, molding, pan coating, solvent dehydration, sonication, spheronization, spray chilling, spray congealing, spray drying, or other processes known in the art. In certain instances, the additive is optionally pre-coated or encapsulated. Suitable additives are optionally utilized to influence the drug release from the composition and/or pharmaceutical dosage form.

Suitable additives utilized in various embodiments described herein include, by way of nonlimiting example, adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, antioxidants, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, co-solvent, opaquants, congealing agents, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, glidants, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, lubricant oils, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, solidifying agent, solvents, solubilizers, spreading agent sweeteners, stabilizers, surface area enhancing agents, suspending agent, thickeners, viscosity increasing agents, waxes and mixtures thereof.

Some non-limiting examples of the hydrophilic or lipophilic additives suitable for the current invention are as follows:

Alcohols and/or Polyols (e.g. ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, glycerol, sorbitol, mannitol, dimethyl isosorbide, polyethylene glycol, fatty acid alcohol, vinyl alcohol polypropylene glycol, polyvinylalcohol, tocopherol s, cellulose cyclodextrins, other derivatives, forms, mixtures thereof, or the like); ethers of polyethylene glycols having an average molecular weight of about 200 to about 20,000 (e.g. tetrahydrofurfuryl alcohol PEG ether, methoxy PEG, or the like); Amides (e.g. 2-pyrrolidone, 2-piperidone, 8- caprolactam, N-alkylpyrrolidone, N-hydroxyalkylpyrrolidone, N-alkylpiperidone, N- alkylcaprolactam, dimethylacetamide, polyvinylpyrrolidone and the like.); Esters (e.g. ethyl propionate, tributylcitrate, acetyl triethylcitrate, acetyl tributyl citrate, triethylcitrate, ethyl oleate, ethyl caprylate, ethyl butyrate, triacetin, propylene glycol monoacetate, propylene glycol diacetate, 8-caprolactone and isomers thereof, 6-valerolactone and isomers thereof, b- butyrolactone and isomers thereof; and other additives known in the art, such as dimethyl acetamide, dimethyl isosorbide, N-methylpyrrolidones, monooctanoin, diethylene glycol monoethyl ether, or the like); Amino acids (e.g. P-aminobenzamidine, sodium glycocholate) mesylate; Amino acids and modified amino acids (e.g. aminoboronic acid derivatives and n- acetylcysteine; Peptides and modified peptides (e.g. bacitracin, phosphinic acid dipeptide derivatives, pepstatin, antipain, leupeptin, chymostatin, elastin, bestatin, phoshporamindon, puromycin, cytochalasin potatocarboxy peptidase inhibitor, amastatin, or the like); Polypeptide protease inhibitors; Mucoadhesive polymers (e.g. polyacrylate derivatives, chitosan, cellulosics, chitosan-EDTA, chitosan-EDTA-antipain, polyacrylic acid, carboxymethyl cellulose etc.); or the like; or combinations thereof.

Some more examples of suitable additives for compositions and/or dosage forms described herein include, by way of non-limiting example, talc, magnesium stearate, silica (e.g. fumed silica, micronized silica, magnesium aluminum silicate etc.) and/or derivatives, polyethylene glycols, surfactants, waxes, oils, cetyl acohol, polyvinyl alcohol, stearic acid, stearic acid salts, stearic acid derivatives, starch, hydrogenated vegetable oils, hydrogenatied castor oils, sodium benzoate, sodium acetate, leucine, PEG, alkyl sulfate salts; acetylated monoglycerides; long- chain alcohols; silicone derivatives; butylated hydroxy toluene (BHT), butylated hydroxyl anisole (BHA), gallic acid, propyl gallate, ascorbic acid, ascorbyl palmitate, 4-hydroxymethyl- 2,6-di-tert-butyl phenol, dry starch, dry sugars, polyvinyl pyrrolidones, starch paste, methacrylic copolymers, bentonite, sucrose, polymericcellulose derivatives, shellac, sugar syrup; com symp; polysaccharides, acacia, tragacanth, guar gum, xanthan gums; alginates; gelatin; gelatin hydrolysate; agar; sucrose; dextrose; PEG, vinyl pyrrolidone copolymers, poloxamers; pregelatinized starch, sorbitol, glucose); acetic acid, hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid and uric acid, vinegar, pharmaceutically acceptable bases, such as an amino acid, an amino acid ester, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium

aluminum silicate, synthetic aluminum silicate, synthetic hydrotalcite, magnesium aluminum hydroxide, diisopropylethylamine, ethanolamine, ethylenediamine, triethanolamine,

triethylamine, triisopropanolamin; salt of a pharmaceutically acceptable cation and an anion; EDTA and EDTA salts; titanium dioxide, food dyes, lakes, natural vegetable colorants, iron oxides, silicates, sulfates, magnesium hydroxide and aluminum hydroxide; halogenated hydrocarbons, trichloroethane, trichloroethylene, dichloromethane, fluorotrichloromethane, diethylether, trehelose, phosphates, citric acid, tartaric acid, gelatin, dextran and mannitol, lactose, mannitol, sodium chloride, potassium chloride, spray-dried lactose, hydrolyzed starches, directly compressible starch, microcrystalline cellulose, cellulosic derivatives, sorbitol, sucrose, sucrose-based materials, calcium sulfate, dibasic calcium phosphate, dextrose, croscarmellose sodium, starch, starch derivatives, clays, gums, cellulose, cellulose derivates, alginates, crosslinked polyvinylpyrrolidone, sodium starch glycolate and microcrystalline cellulose, magnesium oxide, magnesium carbonates; desensitizers, spray-dried flavors, essential oils, ethyl vanillin, styrene/divinyl benzene copolymers, quaternary ammonium compounds, polyethylene glycol, citrate esters (such as triethyl citrate, acetyl triethyl citrate, acetyltributyl citrate), acetylated monoglycerides, glycerin, triacetin, propylene glycol, phthalate esters (e.g., diethyl phthalate, dibutyl phthalate), castor oil, sorbitol and dibutyl sebacate, ascorbic acid, boric acid, sorbic acid, benzoic acid, and salts thereof, parabens, phenols, benzyl alcohol, and quaternary ammonium compounds; alcohols, ketones, esters, chlorinated hydrocarbons water; sweeteners, (e.g. maltose, sucrose, glucose, sorbitol, glycerin and dextrins, aspartame, saccharine, saccharine salts, glycyrrhizin), viscosity modifiers, sugars, polyvinylpyrrolidone, cellulosics, polymers, gums and/or alginates.

Additives can also be materials such as proteins (e.g., collagen, gelatin, Zein, gluten, mussel protein, lipoprotein); carbohydrates (e.g., alginates, carrageenan, cellulose derivatives, pectin, starch, chitosan); gums (e.g., xanthan gum, gum arabic); spermaceti; natural or synthetic waxes; camuaba wax; fatty acids (e.g., stearic acid, hydroxystearic acid); fatty alcohols; sugars; shellacs, such as those based on sugars (e.g., lactose, sucrose, dextrose) or starches; polysaccharide-based shellacs (e.g., maltodextrin and maltodextrin derivatives, dextrates, cyclodextrin and

cyclodextrin derivatives); cellulosic-based polymers (e.g., ethyl cellulose, methyl cellulose, microcrystalline cellulose, sodium carboxymethyl cellulose, hydroxypropylmethyl cellulose, ethyl cellulose, hydroxypropyl cellulose, HPMC acid succinates, cellulose acetate, cellulose nitrate, cellulose acetate butyrate, cellulose acetate trimellitate, carboxymethylethyl cellulose, hydroxypropylmethyl cellulose phthalate), shellacs; inorganics, such as dicalcium phosphate, hydroxyapitite, tricalcium phosphate, talc and titania; polyols, such as mannitol, xylitol and sorbitol; polyethylene glycol esters; and polymers, such as alginates, poly(lactide coglycolide), gelatin, crosslinked gelatin, and agar-agar.

It should be appreciated that there is considerable overlap between the above-listed additives in common usage, since a given hydrophilic or lipophilic additive is often classified differently by different practitioners in the field, or is commonly used for any of several different or overlapping functions. Thus, the above-listed hydrophilic or lipophilic additives should be taken as merely exemplary, and not limiting, of the types of additives that can be included in compositions of the present invention. In certain embodiments, the amounts of such additives are optionally adjusted and/or determined by one skilled in the art, according to the particular properties desired.

In certain embodiments, the at least one pharmaceutically acceptable carrier comprises at least one hydrophilic carrier (e.g., hydrophilic surfactant). In some embodiments, the hydrophilic carrier is a polyoxylated glyceride (e.g., mono-, di-, or tri-glyceride), a polyoxylated vegetable oil, a polyoxylated hydrogenated vegetable oil, a polyoxylated fatty acid (mono-, or di- substituted), combinations thereof, or the tike. In certain embodiments, the at least one pharmaceutically acceptable carrier comprises or further comprises a lipophilic carrier. Lipophilic carriers are selected from, by way of non-limiting example, a lipophilic surfactant, a vegetable oil (e.g., castor oil), a fatty acid, a fatty alcohol, a glyceride (e.g., mono-, di-, or triglyceride), a hydrogenated vegetable oil, a Vitamin E compound (e.g., d,l-a-tocopherol), a trigliceride, a fatty acid, polyoxylated fatty acid, polyoxylated triglyceride, polyoxylated vegetable oil, or combinations thereof. In some embodiments, polyoxylated compounds include polyethoxylated compounds.

In certain embodiments, the at least one hydrophilic carriers make up about 1% to about 99% w/w, about 2% to about 80% w/w, about 2% to about 50% w/w, or about 10% to about 40% w/w of any pharmaceutical composition described herein. In some embodiments, lipophilic carriers make up about 1% w/w to about 99% w/w, about 2% to about 80% w/w, about 10% w/w to about 80% w/w, about 30% w/w, to about 80% w/w, or about 40% to about 80% w/w of any pharmaceutical composition described herein.

In specific embodiments, provided herein is a pharmaceutical composition (e.g., a delayed release dosage form) comprising a hydrophilic carrier. In more specific embodiments, the hydrophilic carrier is or comprises a polyoxylated vegetable oil (e.g., a polyoxylated,

hydrogenated vegetable oil). In still more specific embodiments, a polyoxylated vegetable oil is a polyoxylated castor oil (e.g., a polyoxylated, hydrogenated castor oil). In certain embodiments, the lipidic and/or lipophilic carrier is not a C6-C18 fatty acid. In some embodiments, the lipophilic carrier is a C20+ fatty acid. In some embodiments, the lipidic and/or lipophilic carrier is not a fatty acid or an un-modified (e.g., non-polyoxylated) vegetable oil. In more specific embodiments, the lipidic and/or lipophilic carrier is not oleic acid or castor oil. In certain specific embodiments provided herein is a pharmaceutical composition (e.g., a delayed release dosage form) comprising an amphiphilic carrier. In more specific embodiments, the amphiphilic carrier is or comprises a zwitterionic choline (e.g., phosphatidylcholine). In some specific embodiments, provided herein is a pharmaceutical composition (e.g., a delayed release dosage form) comprising a lipophilic carrier. In more specific embodiments, the lipophilic carrier is or comprises, by way of non-limiting example, a mono-, di- or triglyceride (e.g., glycerol monolinoleate).

In some embodiments, the at least one pharmaceutically acceptable carrier comprises at least one hydrophilic carrier, and at least one lipidic and/or lipophilic carrier. In further embodiments, the at least one pharmaceutically acceptable carrier comprises at least one hydrophilic carrier, at least one lipidic and/or lipophilic carrier, and at least one viscosity enhancer or solidifying agent. In some embodiments, the solidifying agent is a polyethylene glycol (e.g., a high molecular weight polyethylene glycol, such as PEG 8000). In specific embodiments, a pharmaceutical composition described herein comprises, along with a steroidal agent (e.g., a testosterone alkyl ester), a hydrogenated and polyoxylated castor oil and a polyethylene glycol. In more specific embodiments, the pharmaceutical composition comprising a hydrogenated and polyoxylated castor oil and a polyethylene glycol further comprises an additional lipidic and/or lipophilic carrier. In some embodiments, the additional lipidic and/or lipophilic carrier is a monoglyceride, a diglyceride, a Vitamin E compound, or a combination thereof.

In certain embodiments, pharmaceutical compositions described herein include oral dosage forms or delayed release oral dosage forms of any of Tables A to Q. In Tables A to Q, approximate weight percentages of the compositions formulated into the capsules are provided. In specific embodiments, the steroidal compound of any of Capsules A1 to Q2 comprises an alkyl ester testosterone (e.g., testosterone undecanoate). In certain instances, provided in the tables are non-limiting grades and/or sources of components utilized. Disclosure provided in Tables A to Q is not limited to the grades and/or sources described. It is noted that any testosterone ester can be used in place of any specific testosterone ester disclosed in these tables (e.g., testosterone tridecanoate can be used in place of testosterone undecanoate or two testosterone esters can be used e.g., testosterone undecanoate and testosterone tridecanoate).

CR)

Com Glycerides (Maisine 35-1) 50 30-60

NF

Hypromellose (100 cP, K100 5-40 15-25

Premium L V)

It is noted that the above compositions (or elsewhere) can also be prepared without any of the specific solidifying agents and used in the methods described herein.

In certain embodiments, any pharmaceutical composition described herein, e.g., a pharmaceutical composition of any of Tables A to Q can be prepared by (i) combining and heating all ingredients until a molten mixture is obtained (e.g., 50-70° C.); and (ii) encapsulating an amount of molten mixture comprising a select dose (e.g., a therapeutically effective amount or a partial dose of a therapeutically effective amount) of steroidal compound to obtain an oral dosage form. In certain instances, the molten mixture is spray-congealed to obtain beads. In some instances, the molten mixture is sprayed onto inert cores (e.g., sugar spheres) to obtain coated cores. In certain embodiments, such beads, cores, or similar forms are encapsulated or otherwise formulated to provide an oral dosage form. In some instances, the molten mixture is admixed, uniformly dispersed, or granulated over a carrier and compressed into a tablet dosage form. In certain embodiments, prior to compression, the molten mixture/carrier composition is further mixed with one or more pharmaceutical aid including, by way of non-limiting example, glidants, lubricants, binders, or the like. In some embodiments, the carrier is a therapeutically inert carrier such as, by way of non-limiting example, microcrystalline cellulose, starch, lactose, or the like.

In some embodiments, compositions described herein (e.g., compositions set forth in Tables K to M), are optionally filled into a delayed release capsule or shell, or are otherwise coated or encapsulated with a delayed release coat.

Carriers

Provided herein are pharmaceutical compositions comprising a steroidal compound (e.g., one or more testosterone alkyl ester (e.g., testosterone ester), such as testosterone undecanoate) and at least one pharmaceutically acceptable carrier. In certain embodiments, the at least one pharmaceutically acceptable carrier comprises at least one hydrophilic carrier (e.g., hydrophilic surfactant or additive), at least one lipophilic carrier (e.g., lipophilic surfactant or additive), and/or at least one viscosity enhancer or solidifying agent. In specific embodiments, the at least one pharmaceutically acceptable carrier is a hydrophilic carrier. In more specific embodiments, the at least one pharmaceutically acceptable carrier comprises or further comprises a lipophilic carrier. In further embodiments, the at least one pharmaceutically acceptable carrier comprises at least one hydrophilic carrier, at least one lipidic and/or lipophilic carrier, and at least one viscosity enhancer or solidifying agent.

In certain embodiments, hydrophilic carriers include, by way of non-limiting example, a hydrophilic surfactant. In various instances, hydrophilic surfactants are used to provide any one or more of several advantageous characteristics to the compositions, including, by way of non- limiting example: increased solubility of the active ingredient in at least one of the fractions of the carrier that is a solid carrier, improved dissolution of the active ingredient; improved dispersion and/or dissolution of tire lipidic carrier; improved solubilization of the active ingredient upon dissolution; enhanced absorption and-'or bioavailability of the active ingredient, particularly a hydrophilic, hydrophobic, or lipophilic active ingredient; and improved stability, both physical and chemical, of the active ingredient. In various embodiments, the hydrophilic surfactant includes either a single hydrophilic surfactant or a mixture of hydrophilic surfactants. Hydrophilic surfactants also include both ionic or non-ionic surfactants.

In some embodiments, lipophilic carriers include or further include, by way of nonlimiting example, one or more lipophilic surfactant, including one or more lipophilic surfactant, one or more mono-, di-, or triglyceride, or mixtures thereof. In various instances, lipophilic surfactants provide any one or more of the advantageous characteristics listed above for hydrophilic surfactants, and/or enhance the function of other (e.g., hydrophilic) surfactants present in the pharmaceutical composition.

The terms“hydrophilic” and“lipophilic” are relative terms. Hydrophilicity and/or lipophilicity are determined in any manner suitable. In one instances, an empirical parameter is used to characterize the relative hydrophilicity and lipophilicity of the carriers described herein. For example, in one manner, the hydrophilicity and/or lipophilicity non-ionic amphiphilic compounds is the hydrophilic-lipophilic balance (the“HLB” value). Carriers or surfactants with lower HLB values are more lipophilic, and have greater solubility in oils, whereas surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous mediums. This measure is suitable for the surfactants described herein because, generally, surfactants are amphiphilic as they comprise both a polar moiety (e.g., a polar non-charged or charged moiety) and a lipophilic moiety (e.g., an aliphatic group).

Using HLB values as a rough guide, hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as non-ionic, anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable.

Similarly, lipophilic surfactants are compounds having an HLB value less than about 10.

It should be appreciated that the HLB value of a surfactant is merely a rough guide generally used to enable formulation of industrial, pharmaceutical and cosmetic emulsions. For many important surfactants, including several polyethoxylated surfactants, it has been reported that HLB values can differ by as much as about 8 HLB units, depending upon the empirical method chosen to determine the HLB value (Schott, J. Pharm. Sciences, 79(1 ), 87-88 (1990)). Likewise, for certain polypropylene oxide containing block copolymers (poloxamers, available commercially as PLURONIC® surfactants, BASF Corp.), the HLB values are not always authoritative indicators of the true physical chemical nature of the compounds. Finally, commercial surfactant products are generally not pure compounds, but are often complex mixtures of compounds, and tire HLB value reported for a particular compound may more accurately be characteristic of the commercial product of w'hich the compound is a major component. Different commercial products having tire same primary surfactant component can, and typically do, have different HLB values. In addition, a certain amount of lot-to-lot variability is expected even for a single commercial surfactant product. Thus, keeping these considerations involved, a person of ordinary skill in the art is able to utilize HLB values and the identity of a given product to determine surfactants for suitable lipophilicity and/or hydrophilicity for use in the pharmaceutical compositions described herein.

As used herein, useful surfactants include any surfactant that is pharmaceutically acceptable and is suitable for use in a pharmaceutical composition. Suitable surfactants include anionic, cationic, zwitterionic and non-ionic surfactants. Provided herein (e.g., in the Tables) are several general classes of surfactants. The HLB values given in the Tables below generally represent the HLB value as reported by the manufacturer of the corresponding commercial product. In cases where more than one commercial product is listed, the HLB value in the Tables is the value as reported for one of the commercial products, a rough average of the reported values, or a value that, in the judgment of the present inventors, is more reliable.

Surfactants described in the Tables are illustrative and are provided as non-limiting examples. For example, refined, distilled or fractionated surfactants, purified fractions thereof, or re-esterified fractions, are also within the scope of surfactants described herein, although they are not specifically listed in the Tables.

In some embodiments, surfactants described herein include polyoxylated fatty acids, such as polyethoxylated fatty acids (i.e., PEC-fatty acid esters). Provided in Table 1 is a list of illustrative and non-limiting examples of polyethoxylated fatty acid monoester surfactants.

B

Furthermore, in some embodiments, surfactants described herein include, by way of non-limiting example, polyethylene glycol (PEG) fatty acid diesters. Illustrative and non-limiting examples of PEG-fatty acid diesters are shown in Table 2.

As discussed above, in some embodiments, pharmaceutical compositions described herein comprise mixtures of surfactants, including, e.g., mixtures of two or more commercial surfactant products. Several PEG-fatty acid esters are marketed commercially as mixtures or mono- and diesters. Illustrative and non-limiting examples of surfactant mixtures are shown in Table 3.

In some embodiments, surfactants described herein include, by way of non-limiting example, polyethylene glycol glycerol fatty acid esters (PEG glycerol fatty acid esters). Illustrative and non-limiting examples of PEG glycerol fatty acid esters are shown in Table 4.

In certain embodiments, surfactants of different degrees of iipophilicity or hydrophilicity are prepared by reaction of alcohols or polyalcohols with a variety of natural and/or hydrogenated oils. In some embodiments, the oils used are castor oil or hydrogenated castor oil or an edible vegetable oil such as com oil, olive oil, peanut oil, palm kernel oil, apricot kernel oil, or almond oil. In specific embodiments, alcohols include glycerol, propylene glycol, ethylene glycol, polyethylene glycol, sorbitol, and pentaerythritol. In certain embodiments, such surfactants are utilized in the pharmaceutical compositions described herein. Illustrative and non-limiting examples of surfactants of this class suitable for use in the pharmaceutical compositions described herein are show in Table 5.

TABLE 5

kernel oil

PEG-6 olive oil Labrafil ® M 1980 CS (Gattefosse) 4

PEG-6 peanut oil Labrafil ® M 1969 CS (Gattefosse) 4 PEG-6 hydrogenated Labrafil ® M 2130 BS (Gattefosse) 4 palm

kernel oil

PEG-6 palm kernel oil Labrafil ® M 2130 CS (Galtefosse) 4 PEG-6 triolein Labrafil ® M 2735 CS (Gatlefbsse) 4 PEG-8 corn oil Labrafil ® WL 2609 BS (Gattefosse) 6-7 PEG-20 com Crovol M40 (Croda) 10 glycerides

PEG-20 almond Crovol A40 (Croda) 10 glycerides

PEG-25 trioleate TAGAT® TO (Goldschmidt) 11 PEG-40 palm Crovol PK-70 >10 kernel oil

PEG-60 com Crovol M70(Croda) 15 glycerides

PEG-60 almond Crovol A70 (Croda) 15 glycerides

PEG-4 caprylic/capric Labrafac® Hydro (Gattefosse), 4-5 triglyceride

PEG-8 caprylic/capric Labrasol (Gattefosse), >10 glycerides Labrafac CM 10 (Gattefosse)

PEG-6 caprylic/capric SOFTIGEN ® 767 (Huls). 19 glycerides Glycerox 767 (Croda)

Lauroyl macrogol-32 GELUCIRE 44/14 (Gattefosse) 14 glyceride

Stearoyl macrogol GELUCIRE 50/13 (Gattefosse) 13 glyceride

Mono, di, tri, tetra SorbitoGlyceride (Gattefosse) <10 esters of

vegetable oils

and sorbitol

Pentaerythrityl Crodamol PTIS (Croda) <10 tetraisostearate

Pentaerythrityl Albunol DS (Taiwan Surf.) <10 distearate

Pentaerythrityl Liponate PCM (Lipo Chem.) <10 tetraoleate

Pentaerythrityl Liponate PS-4 (Lipo Chem.) <10 tetrastearate

Pentaerythrityl Liponate PE-810 (Lipo Chem.), <10

In some embodiments, surfactants utilized in the pharmaceutical compositions described herein include, by way of non-limiting example, polyglycerized fatty acids. Illustrative and non-limiting examples of suitable polyglyceryl esters are shown in Table 6.

As discussed above, mixtures of surfactants are also used, in some embodiments, in the pharmaceutical compositions described herein. Mixtures of surfactants include, by way of nonlimiting example, mixtures of propylene glycol fatty acid esters and glycerol fatty acid esters are suitable and are commercially available. Illustrative and non-limiting examples of such mixtures of surfactants include, by way of non-limiting example, those shown in Table 8.

TABLE 8

In certain embodiments, an important class of surfactants includes the class of mono- and diglycerides. These surfactants are generally lipophilic. Illustrative and non-limiting examples of these surfactants are given in Table 9.

In some embodiments, surfactants utilized in the pharmaceutical compositions described herein include sterols and derivatives of sterols. In various embodiments, these surfactants are hydrophilic or lipophilic. Illustrative and non-limiting examples of surfactants of this class are shown in Table 10.

In some embodiments, surfactants useful in the pharmaceutical compositions described herein include a variety ofPEG-sorbitan fatty acid esters. In general, these surfactants are hydrophilic, although several lipophilic surfactants of this class can be used. Illustrative and non-limiting examples of these surfactants are shown in Table 11.

In some embodiments, surfactants utilized herein include ethers of polyethylene glycol and alkyl alcohols. Illustrative and non-limiting examples of these surfactants are shown in Table 12.

In certain embodiments, surfactants utilized in the pharmaceutical compositions described herein include esters of sugars. Illustrative and non-limiting examples of such surfactants are shown in Table 13.

In some embodiments, surfactants utilized in the pharmaceutical compositions described herein include polyethylene glycol alkyl phenols, e.g., hydrophilic PEG-alkyl phenol surfactants.

Illustrative and non-limiting examples of these surfactants are shown in Table 14.

In certain embodiments, surfactants utilized in pharmaceutical compositions described herein include polyoxyethylene-poly oxy propylene block copolymers. POE-POP block copolymers are a unique class of polymeric surfactants. The unique structure of the surfactants, with hydrophilic POE and lipophilic POP moieties in well-defined ratios and positions, provides a wide variety of surfactants suitable for use in the present invention. These surfactants are available under various trade names, including Synperonic PE series (ICI), Pluronic® series (BASF), Emkalyx, Lutrol (BASF), Supronic, Monoian, Pluracare, and Plurodac. The generic term for these polymers is “poloxamer” (CAS 9003-1 1-6). These polymers have the formula:

H0(C2H4())a(C3H60>(C2H4C))J [; wherein the terms“a” and“b” denote the number of polyoxyethylene and polyoxypropylene units, respectively.

Illustrative and non-limiting examples of suitable surfactants of this class are shown in Table 15. Since the compounds are widely available, commercial sources are not listed in the Table. The compounds are listed by generic name, with the corresponding“a" and“b” values.

In some embodiments, surfactants utilized in pharmaceutical compositions described herein include sorbitan esters of fatty acids. Illustrative and non-limiting examples of such surfactants are shown in Table 16.

In certain embodiments, surfactants utilized in pharmaceutical compositions described herein include esters of lower alcohols (C2 to Gi) and fatty acids (Csto Cis). Illustrative and nonlimiting examples of these surfactants are shown in Table 17.

In some embodiments, hydrophilic surfactants utilized in pharmaceutical compositions described herein include ionic surfactants (e.g., cationic, anionic and zwitterionic surfactants). In specific embodiments, anionic surfactants include fatty acid salts and bile acid salts. In certain specific embodiments, cationic surfactants include carnitines. In some specific embodiments, ionic surfactants include, by way of non-limiting example, sodium oleate, sodium lauryl sulfate, sodium lauryl sarcosinate, sodium dioctyl sulfosuccinate, sodium cholate, sodium taurocholate; lauroyl carnitine; palmitoyl carnitine; and myristoyl carnitine. Illustrative and non-limiting examples of such surfactants are shown in Table 18. For simplicity, exemplary counterions are shown in the entries in the Table. In various embodiments, such counterions are optionally substituted with any suitable counterion. For example, although the fatty acids are shown as sodium salts, other cation counterions are optionally used, such as alkali metal cations or ammonium. Unlike certain non-ionic surfactants, these ionic surfactants are generally available as pure compounds, rather than commercial (proprietary) mixtures. Because these compounds are readily available from a variety of commercial suppliers, such as Aldrich, Sigma, and the like, commercial sources are not generally listed in the Table.

In some embodiments, surfactants utilized in pharmaceutical compositions described herein include ionizable surfactants. In certain embodiments, ionizable surfactants, when present in their unionized (neutral, non-salt) form, are lipophilic surfactants suitable for use in the compositions of the present invention. Particular examples of such surfactants include free fatty acids, particularly C6-C22 fatty acids, and bile acids. More specifically, suitable unionized ionizable surfactants include the free fatty acid and bile acid forms of any of the fatty acid salts and bile salts shown in Table 18.

In some instances, derivatives of oil -soluble vitamins, such as vitamins A, D, E, K, etc., are also useful surfactants for use in the pharmaceutical compositions described herein. An example of such a derivative is tocopheryl PEG-1000 succinate (TPGS, available from Eastman).

In specific embodiments, surfactants or mixtures of surfactants that solidify (e.g., form a solid, a semi-solid, a gel, a jelly, a paste, or the like) at ambient room temperature are utilized in the pharmaceutical compositions described herein. In certain specific embodiments, surfactants or mixtures of surfactants utilized in the pharmaceutical compositions described herein solidify (e.g., form a solid, a semi-solid, a gel, a jelly, a paste, or the like) at ambient room temperature when combined with additional agents (e.g., particular lipophilic components, such as triglycerides, vitamins (e.g., Vitamin E), or the like, viscosity modifiers, stabilizers, solidifying agents, binders, thickeners, or the like). Such additional agents are optionally utilized in the pharmaceutical compositions described herein. In certain embodiments, pharmaceutical compositions described herein comprise a hydrophilic carrier (e.g., a hydrophilic surfactant), a lipophilic carrier, and/or a viscosity modifier or solidifying agent.

In some specific embodiments, non-ionic hydrophilic surfactants include alkylglucosides;

alkylmaltosides; alkylthioglucosides, lauryl macrogolglycerides; polyoxyethylene alkyl ethers; polyoxyethylene alkylphenols; polyethylene glycol fatty acids esters; polyethylene glycol glycerol fatty acid esters; polyoxyethylene sorbitan fatty acid esters, polyoxyethylene- poly oxypropylene block copolymers; polyglycerol fatty acid esters; polyoxyethylene glycerides; polyoxyethylene sterols, derivatives, and analogues thereof, polyoxyethylene vegetable oils, polyoxyethylene hydrogenated vegetable oils; reaction mixtures of polyols with fatty acids, glycerides, vegetable oils, hydrogenated vegetable oils, and sterols; sugar esters, sugar ethers; sucroglycerides; polyethoxylated fat-soluble vitamins or derivatives; and mixtures thereof.

In certain specific embodiments, the non-ionic hydrophilic surfactant is selected from, by way of non-limiting example, polyoxyethylene alkylethers; polyethylene glycol fatty acids esters;

polyethylene glycol glycerol fatty acid esters; polyoxyethylene sorbitan fatty acid esters; polyoxyethylene-polyoxypropylene block copolymers; polyglyceryl fatty acid esters; polyoxyethylene glycerides; polyoxyethylene vegetable oils, and polyoxyethylene hydrogenated vegetable oils. In various embodiments, the glyceride is a monoglyceride, diglyceride, triglyceride, or a mixture thereof. In some specific embodiments, non-ionic hydrophilic surfactants are the products of reaction mixtures of polyols and fatty acids, glycerides, vegetable oils, hydrogenated vegetable oils or sterols. These reaction mixtures are largely composed of the transesterification products of the reaction, along with often complex mixtures of other reaction products. In more specific embodiments, the polyol is glycerol, ethylene glycol, polyethylene glycol, sorbitol, propylene glycol, pentaerythritol, or a saccharide.

In certain specific embodiments, the hydrophilic surfactant is or includes an ionic surfactant. Specific ionic surfactants include alkyl ammonium salts; bile acids and salts, analogues, and derivatives thereof; fusidic acid and derivatives thereof; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids, oligopeptides, and polypeptides; acyl lactylates; mono-,diacetylated tartaric acid esters of mono-, diglycerides; succinylated monoglycerides; citric acid esters of mono-, diglycerides; alginate salts; propylene glycol alginate; lecithins and hydrogenated lecithins; lysolecithin and hydrogenated

lysolecithins; lysophospholipids and derivatives thereof; phospholipids and derivatives thereof; salts of alkylsulfates; salts of fatty acids; sodium docusate; carnitines; and mixtures thereof. In some specific embodiments, ionic surfactants include bile acids and salts, analogues, and derivatives thereof; lecithins, lysolecithin, phospholipids, lysophospholipids and derivatives thereof; salts of alkylsulfates; salts of fatty acids; sodium docusate; acyl lactylates; mono- .diacetylated tartaric acid esters of mono-, di glycerides; succinylated monoglycerides; citric acid esters of mono-diglycerides; carnitines; and mixtures thereof. In more specific embodiments, ionic surfactants include, by way of non-limiting example, lecithin, lysolecithin,

phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidic acid, phosphatidylserine, lysophosphatidylcholine, lysophosphatidylethanolamine,

lysophosphatidylglycerol, lysophosphatidic acid, lysophosphatidylserine, PEG- phosphatidylethanol amine, PVP-phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/digly ceri des, cholate, taurocholate, glycocholate, deoxycholate, taurodeoxycholate, chenodeoxycholate, glycodeoxycholate, glycochenodeoxycholate, taurochenodeoxy cholate, ursodeoxycholate, tauroursodeoxycholate, glycoursodeoxy cholate, cholylsarcosine, N-methyl taurocholate, caproate, capiylate, caprate, laurate, myristate, palmitate, oleate, ricinoleate, linoleate, linolenate, stearate, lauryl sulfate, teracecyl sulfate, docusate, lauroyl carnitines, palmitoyl carnitines, myristoyl carnitines, and salts and mixtures thereof. In more specific embodiments, ionic surfactants are selected from lecithin, lysolecithin, phosphatidylcholine, phosphatidylethanol amine, phosphatidylglycerol,

lysophosphatidylcholine, PEG-phosphati dyl ethanolamine, lactylic esters of fatty acids, stearoyl - 2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholate, taurocholate, glycocholate, deoxycholate, taurodeoxycholate, glycodeoxycholate, cholylsarcosine, caproate, capiylate, caprate, laurate, oleate, lauryl sulfate, docusate, and salts and mixtures thereof, with the most preferred ionic surfactants being lecithin, lactylic esters of fatty' acids, stearoyl-2- lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, taurocholate, capiylate, caprate, oleate, I amyl sulfate, docusate, and salts and mixtures thereof.

In various embodiments, lipophilic surfactants are selected from, by way of non-limiting example, alcohols; polyoxyethylene alkylethers; fatty acids; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; polyethylene glycol fatty acids esters; polyethylene glycol glycerol fatty acid esters; polypropylene glycol fatty acid esters;

polyoxyethylene glycerides; lactic acid derivatives of mono/di glycerides; propylene glycol diglycerides; sorbitan fatty acid esters; polyoxyethylene sorbitan fatty acid esters;

polyoxyethylene-poly oxypropy 1 ene block copolymers; transesterified vegetable oils; sterols; sterol derivatives; sugar esters; sugar ethers; sucroglycerides; polyoxyethylene vegetable oils; and polyoxyethylene hydrogenated vegetable oils. As with the hydrophilic surfactants, lipophilic surfactants are optionally the products of reaction mixtures of polyols and fatty acids, glycerides, vegetable oils, hydrogenated vegetable oils, and sterols. In specific embodiments, lipophilic surfactants are selected from fatty acids; lower alcohol fatty acid esters; polyethylene glycol glycerol fatty acid esters; polypropylene glycol fatty acid esters; polyoxyethylene glycerides; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lactic acid derivatives of mono/diglycerides; sorbitan fatty acid esters; polyoxyethylene sorbitan fatty acid esters;

polyoxyethylene-polyoxypropylene block copolymers; polyoxyethylene vegetable oils;

polyoxyethylene hydrogenated vegetable oils; and reaction mixtures of polyols and fatty acids, glycerides, vegetable oils, hydrogenated vegetable oils, and sterols. In certain specific embodiments, lipophilic surfactants are selected from lower alcohol fatty acids esters;

polypropylene glycol fatty acid esters; propylene glycol fatty acid esters; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lactic acid derivatives of mono/diglycerides; sorbitan fatty acid esters; polyoxyethylene vegetable oils; and mixtures thereof, with glycerol fatty acid esters and acetylated glycerol fatty acid esters being most preferred. Among the glycerol fatty acid esters, the esters are, e.g., mono- or diglycerides, or mixtures of mono- and diglycerides, where the fatty acid moiety is a Ceto C22 fatty acid. In some specific embodiments, lipophilic surfactants are selected from the products of reaction mixture of polyols and fatty acids, glycerides, vegetable oils, hydrogenated vegetable oils, and sterols. In more specific

embodiments, polyols are polyethylene glycol, sorbitol, propylene glycol, and pentaeiythritol. In certain embodiments, pharmaceutical compositions described herein include a lipophilic component or carrier. In some embodiments, the lipophilic carrier is selected from lipophilic surfactants, triglycerides, and Vitamin E compounds (e.g., d,l-a-tocopherol). In specific embodiments, triglycerides utilized in the pharmaceutical compositions described herein are those that solidify (e.g., form a solid, a semi-solid, a gel, a jelly, a paste, or the like) at ambient room temperature, with or without addition of appropriate additives, or those which in combination with particular surfactants and/or active ingredients solidify at room temperature. Illustrative and non-limiting examples examples of triglycerides suitable for use in the pharmaceutical compositions described herein are shown in Table 19. In general, these triglycerides are readily available from commercial sources. For several triglycerides, representative commercial products and/or commercial suppliers are listed.

In certain embodiments, the triglycerides utilized in the pharmaceutical compositions described herein include fractionated triglycerides, modified triglycerides, synthetic triglycerides, and mixtures of triglycerides are also within the scope of the invention. In specific embodiments, triglycerides include, by way of non-limiting example, vegetable oils, fish oils, animal fats, hydrogenated vegetable oils, partially hydrogenated vegetable oils, medium and long-chain triglycerides, and structured triglycerides. It should be appreciated that several commercial surfactant compositions contain small to moderate amounts of triglycerides, typically as a result of incomplete reaction of a triglyceride starting material in, for example, a transesterification reaction. Such commercial surfactant compositions, while nominally referred to as“surfactants”, may be suitable to provide all or pan of the triglyceride component for the compositions of the present invention. Examples of commercial surfactant compositions containing triglycerides include some members of the surfactant families Gelucires (Gattefosse), Maisines (Gattefosse), and Imwitors (Hiils). Specific examples of these compositions are: Gelucire 44/14 (saturated polyglycolized glycerides); Gelucire 50/13 (saturated polyglycolized glycerides); Gelucire 53/10 (saturated polyglycolized glycerides); Gelucire 33/01 (semi-synthetic triglycerides of Cs- Cissaturated fatty acids); Gelucire 39/01 (semi-synthetic glycerides); other Gelucires, such as 37/06, 43/01, 35/10, 37/02, 46/07, 48/09, 50/02, 62/05, or the like; Maisine 35-1 (linoleic glycerides); and Imwitor 742 (capiylic/capric glycerides).

Additional Agents The pharmaceutical compositions described herein optionally include one or more additional agents or additives. In certain instances, suitable additives include those that facilitate

formulating a pharmaceutical composition described herein as an oral dosage form and include, e.g., coatings and capsule components. Further additives include, by way of non-limiting example, solubilizers, enzyme inhibitors, anti-foaming agents, antioxidants, binders, buffering agents, chelating agents, diluents, disintegrants, flavoring agents, preservatives, sweeteners, thickeners, or the like.

In some embodiments, pharmaceutical compositions provided herein optionally include one or more solubilizers, i.e., additives to increase the solubility of the pharmaceutical active ingredient or other composition components in the solid carrier. Suitable solubilizers for use in the compositions of the present invention include: alcohols, polyols, ethers of polyethylene glycols, amides, esters or the like. Alcohols and polyols include, by way of non-limiting example, ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and other cellulose derivatives, cyelodextrins and cyclodextrin derivatives. Ethers of polyethylene glycols include those having an average molecular weight of about 200 to about 6000, such as, by way of non-limiting example, tetrahydrofurfmyl alcohol PEG ether (glycofurol, available commercially from BASF under the trade name Tetraglycol) and methoxy PEG (Union Carbide). Amides include, by way of non-limiting example, 2-pyrrolidone, 2-piperidone, e-caprolactam, N- alkylpyrrolidone, N-hydroxyalkylpyrrolidone, N-alkylpiperidone, N-alkylcaprolactam, dimethylacetamide, and polyvinylpyrrolidone. Esters include, by way of non-limiting example, ethyl propionate, tri butyl citrate, acetyl triethylcitrate, acetyl tributyl citrate, triethylcitrate, ethyl oleate, ethyl caprylate, ethyl butyrate, triacetin, propylene glycol monoacetate, propylene glycol di acetate, e-caprolactone and isomers thereof, d-valerolactone and isomers thereof, b- butyrolactone and isomers thereof. Other solubilizers include, by way of non-limiting example, dimethyl acetamide, dimethyl isosorbide (Arlasolve DMi (ICT)), N-methylpyrrolidones

(Pharmasolve (ISP)), monooctanoin, diethylene glycol monoethyl ether (available from

Gattefosse under the trade name Transcutol), and water. Mixtures of solubilizers are also within the scope of the present disclosure. ELxcept as indicated, these compounds are readily available from standard commercial sources. In specific embodiments, solubilizers include, by way of non-limiting example, triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N-methy 1 py rroli done, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropyl methyl cellulose, hydroxypropyl cyclodextrins, ethanol, polyethylene glycol 200-600, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide. In certain specific embodiments, solubilizers include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and propylene glycol. The amount of solubilizer included in the pharmaceutical compositions described herein is any suitable amount.

Anti-adherents (anti-sticking agents, glidants, flow promoters, lubricants) include, by way of non-limiting example, talc, magnesium stearate, fumed silica (Carbosil, Aerosil), micronized silica (Syloid No. FP 244, Grace U.S.A.), polyethylene glycols, surfactants, waxes, stearic acid, stearic acid salts, stearic acid derivatives, starch, hydrogenated vegetable oils, sodium benzoate, sodium acetate, leucine, PEG-4000 and magnesium lauryl sulfate. Antioxidants include, by way of non-limiting example, BHT, BHA, gallic acid, propyl gallate, ascorbic acid, ascorbyl palmitate, 4-hydroxym ethyl-2, 6-di-tert-butyl phenol, and tocopherol. Binders (adhesives), i.e., agents that impart cohesive properties to powdered materials through particle-particle bonding, include, by way of non-limiting example, matrix binders (dry' starch, dry' sugars), film binders (PVP, starch paste, celluloses, bentonite, sucrose), and chemical binders (polymeric cellulose derivatives, such as carboxy methyl cellulose, HPC and HPMC; sugar syrups; com syrup; water soluble polysaccharides such as acacia, tragacanth, guar and alginates; gelatin; gelatin

hydrolysate; agar; sucrose; dextrose; and non-cellulosic binders, such as PVT, PEG, vinyl pyrrolidone copolymers, pregelatinized starch, sorbitol, and glucose). Buffering agents, include an acid and a base, wherein the acid is a pharmaceutically acceptable acid, such as hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p- toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid and uric acid, and the base is a pharmaceutically acceptable base, such as an amino acid, an amino acid ester, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium aluminum silicate, synthetic aluminum silicate, synthetic hydrotalcite, magnesium aluminum hydroxide, diisopropylethyl amine, ethanolamine, ethylenediamine, triethanolamine, triethyl amine, triisopropanolamine, or a salt of a

pharmaceutically acceptable cation and acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, an amino acid, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, a fatty acid, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para- bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesul fonic acid, and uric acid. Chelating agents include, by way of non-limiting example, EDTA and EDTA salts. Colorants or opaquants include, by way of non-limiting example, titanium dioxide, food dyes, lakes, natural vegetable colorants, iron oxides, silicates, sulfates, magnesium hydroxide and aluminum hydroxide. Diluents or fillers include, by way of non-limiting example, lactose, mannitol, talc, magnesium stearate, sodium chloride, potassium chloride, citric acid, spray-dried lactose, hydrolyzed starches, directly compressible starch, microcrystalline cellulose, cellulosics, sorbitol, sucrose, sucrose-based materials, calcium sulfate, dibasic calcium phosphate and dextrose. Disintegrants and super disintegrants include, by way of non-limiting example, croscarmellose sodium, starch, starch derivatives, clays, gums, cellulose, cellulose derivatives, alginates, crosslinked polyvinypyrrolidone, sodium starch glycolate and microcrystalline cellulose. Flavorants or desensitizers include, by way of non-limiting example, spray-dried flavors, essential oils and ethyl vanillin. Plasticizers include, by way of non-limiting example, polyethylene glycol, citrate esters (e.g., triethyl citrate, acetyl triethyl citrate, acetyltributyl citrate), acetylated monoglycerides, glycerin, triacetin, propylene glycol, phthalate esters (e.g., diethyl phthalate, dibutyl phthalate), castor oil, sorbitol and dibutyl seccate. Preservatives include, by way of non-limiting example, ascorbic acid, boric acid, sorbic acid, benzoic acid, and salts thereof, parabens, phenols, benzyl alcohol, and quaternary ammonium compounds. Solvents include, by way of non-limiting example, alcohols, ketones, esters, chlorinated hydrocarbons and water. Sweeteners include, by way of non-limiting example, natural sweeteners such as maltose, sucrose, glucose, sorbitol, glycerin and dextrins, and artificial sweeteners, such as aspartame, saccharine and saccharine salts. Thickeners (viscosity modifiers, thickening agents) include, by way of non-limiting example, sugars, polyvinylpyrrolidone, cellulosics, polymers, high molecular weight polyethylene glycols (e.g., PEG 8000), and alginates. Additives also include, by way of non-limiting example, proteins (e.g., collagen, gelatin, Zein, gluten, mussel protein, lipoprotein); carbohydrates (e.g., alginates, carrageenan, cellulose derivatives, pectin, starch, chitosan); gums (e.g., xanthan gum, gum arabic); spermaceti; natural or synthetic waxes;

camuaba wax; fatty acids (e.g., stearic acid, hydroxy stearic acid); fatty alcohols; sugars, shellacs, such as those based on sugars (e.g., lactose, sucrose, dextrose) or starches; polysaccharide-based shellacs (e.g., maltodextrin and maltodextrin derivatives, dextrates, cyclodextrin and

cyclodextrin derivatives); cellulosic-based shellacs (e.g., microcrystalline cellulose, sodium carboxymethyl cellulose, hydroxypropylmethyl cellulose, ethyl cellulose, hydroxypropyl cellulose, cellulose acetate, cellulose nitrate, cellulose acetate butyrate, cellulose acetate trimellitate, carboxymethylethyl cellulose, hydroxypropylmethyl cellulose phthalate); inorganics, such as dicalcium phosphate, hydroxyapitite, tricalcium phosphate, talc and titania; polyols, such as mannitol, xylitol and sorbitol; polyethylene glycol esters; and polymers, such as alginates, poly(lactide coglycolide), gelatin, crosslinked gelatin, and agar-agar.

It should be appreciated that there is considerable overlap between the above-listed additives in common usage, since a given additive is often classified differently by different practitioners in the field, or is commonly used for any of several different functions. Thus, the above-listed additives should be taken as merely exemplary', and not limiting, of the types of additives that can be included in compositions of the present invention. The amounts of such additives can be readily determined by one skilled in the art, according to the particular properties desired.

The compositions and unit dosage forms can be prepared by any suitable method known to the skilled artisan or developed in view of the teachings herein.

In one specific aspect, the cameras) and API are brought to or maintained at a temperature at which they are flowable (e.g., above 10 °C, 20 °C, 25 °C, 30 °C, 35 °C, or 40 °C). In one aspect, the mixture of carrier and API is a clear solution at a specified temperature (e.g., above 10 °C,

20 °C, 25 °C, 30 °C, 35 °C, or 40 °C). In one aspect, the mixture of carrier and API is a cloudy or hazy solution at a specified temperature (e.g., below 10 °C, 20 °C, 25 °C, 30 °C, 35 °C, or 40 °C).

In one example, the composition is prepared by weighing all of the components, except the API into a clean stainless steel container and mixed together at ambient temperature or at elevated temperatures e.g., at about 25°C to about 30°C, at about 30°C to about 35°C, at about 35°C to about 40°C, at about 40°C to about 45°C, at about 45°C to about 45°C, or 50°C to about 70°C, using a stirrer. The API is added and stirred into the mixture of other components until the API dissolves. A predetermined quantity of this“liquid fill material” is disposed into a capsule (for example, hard gelatin capsule) to get the required API dose per dosage unit. The capsules are allowed to cool at room temperature, banded (if required) and packaged in a HOPE bottle and tightly closed with an appropriate lid. It is noted that various capsule sizes (e.g., hard gel or soft gel) are available to the skilled artisan and allow for variations in the amount of loading of API in mg per unit dosage form. Typically, soft gel capsules for oral administration have fill volumes of less than 1.5 mL, 1.3 mL or 1.25 mL with numerous incremental fill volumes in these ranges. Similarly, hard gel capsules typically have fill volumes of less than 1.25 mL, 1.10 mL or 1 mL. Due to the nature of some hard gel capsules, the total fill volume may not be useable. There is a practical limit on the temperature at which capsules can be filled - for example temperature above 40 °C typically melt, deform, or otherwise damage soft gel capsules typically employed in the industry. Hard gel capsules are typically less sensitive to temperature and can be filled at higher temperatures e.g., above 40 °C.

In certain embodiments, any pharmaceutical composition described herein, e.g., a can be prepared by (i) combining and heating all ingredients until a molten mixture is obtained (e.g., 50- 70°C); and (ii) encapsulating an amount of molten mixture comprising a select dose (e.g., a therapeutically effective amount or a partial dose of a therapeutically effective amount) API to obtain an oral dosage form. In certain instances, the molten mixture is spray-congealed to obtain beads. In some instances, the molten mixture is sprayed onto inert cores (e.g., sugar spheres) to obtain coated cores. In certain embodiments, such beads, cores, or similar forms are encapsulated or otherwise formulated to provide an oral dosage form. In some instances, the molten mixture is admixed, uniformly dispersed, or granulated over a carrier and compressed into a tablet dosage form. In certain embodiments, prior to compression, the molten mixture/carrier composition is further mixed with one or more pharmaceutical aid including, by way of non-limiting example, glidants, lubricants, binders, or the like. In some embodiments, the carrier is a therapeutically inert carrier such as, by way of non-limiting example, microcrystalline cellulose, starch, lactose, or the like.

In various embodiments, pharmaceutical compositions described herein are formulated as oral dosage forms. Oral dosage forms are prepared by any suitable process including one or more steps of, by way of non-limiting example, agglomeration, air suspension chilling, air suspension drying, balling, coacervation, comminution, compression, pelletization, cryopelletization, encapsulation, extrusion, granulation, homogenization, inclusion complexation, lyophilization, nanoencapsulation, melting, mixing, molding, pan coating, solvent dehydration, sonication, spheronization, spray chilling, spray congealing, spray drying, or the like.

In some embodiments, a pharmaceutical composition described herein is formulated with a substrate to form an oral dosage form. In various embodiments, substrates useful for formulating pharmaceutical compositions described herein as oral dosage forms include or comprise, by way of non-limiting example, a powder or a multiparticulate (e.g., one or more granule, one or more pellet, one or more bead, one or more spherule, one or more beadlet, one or more microcapsule, one or more millisphere, one or more mini capsule, one or more microcapsule, one or more nanocapsule, one or more nanosphere, one or more microsphere, one or more minitablet, one or more tablet, one or more capsule, or one or more combinations thereof). In certain instances, a powder constitutes a finely divided (milled, micronized, nanosized, precipitated) form of an active ingredient or additive molecular aggregates or a compound aggregate of multiple components or a physical mixture of aggregates of an active ingredient and/or additives.

Exemplary Formulation Embodiments

Provided in this section are formulations for use in the methods described herein. It is noted that any testosterone ester can used in place of the specified API (e.g., testosterone undecanoate or a combination of testosterone undecanoate and testosterone tridecanoate).

Table 1A. Drag + Carriers

000 r, 60. 07. idone, on ntiids, ants, pH ole ol 000, ate, er XIII Carrier II. Compositions composed of solubilizer (Propylene glycol mono or di-caprylate), hydrophilic additives, and other additives for Composition A to E

Carrier IX. Compositions composed of solubilizer ((9Z,12Z)-9,12-Octadecadienoic acid), hydrophilic additives, and other additives for Composition A to E

Shown below are various compositions suitable for oral administration as described herein. In these Examples the amount of excipient adds up to 100% (does not include the API) and the API weight percent is the final weight percent in the pharmaceutical composition.

The API in these compositions in specific compositions is testosterone tridecanoate, testosterone undecanoate, or a combination thereof. Alternatively, the skilled artisan understands that any other testosterone ester or steroid, or steroid ester can be used in these compositions accordingly. Excipient 1 in specific compositions is (9Z)-octadec-9-enoic acid. Excipient 2 in specific compositions is a combination of mono-, di-, or tri- propane-1, 2, 3-triol esters of octadecanoic acid and hexadecanoic acid; H-(0-CH2-CH2)n-0H where n is an integer from 3 to 900;

octadecanoic acid; (lR,2S,5R)-2-isopropyl-5-methylcyclohexanol or a combination of one or more of (lR,2S,5R)-2-isopropyl-5-methylcyclohexanol, (2S,5R)-2-Isopropyl-5- methylcyclohexanone, Acetic acid [(lR,2S,5R)-2-isopropyl-5-methylcyclohexyl] ester, 1,3,3- Trimethyl-2-oxabicyclo[2,2,2]octane, and (R)-l-methyl-4-(l-methylethenyl)cyclohexene; or a combination thereof. Excipient 3 in specific compositions is a polyoxylated hydrogenated vegetable oil. Excipient 4 in specific compositions is ascorbyl palmitate. These compositions can be filled into soft gel or hard gel capsules depending on its flowability at the temperatures useful for making these dosage forms. Examplary Compositions

Shown below are various compositions suitable for oral administration as described herein. In these Examples the amount of excipient adds up to 100% (does not include the API) and the API weight percent is the final weight percent in the pharmaceutical composition.

The API in these compositions in specific compositions is testosterone tridecanoate, testosterone undecanoate, or a combination thereof. Alternatively, the skilled artisan understands that any other testosterone ester or steroid, or steroid ester can be used in these compositions accordingly. Excipient 1 in specific compositions is (9Z)-octadec-9-enoic acid. Excipient 2 in specific compositions is a combination of mono-, di-, or tri- propane-1, 2, 3-triol esters of octadecanoic acid and hexadecanoic acid, octadecanoic acid or a combination thereof. Excipient 3 in specific compositions is (lR,2S,5R)-2-isopropyl-5-methylcyclohexanol or a combination of one or more of (lR,2S,5R)-2-isopropyl-5-methylcyclohexanol, (2S,5R)-2-Isopropyl-5-methylcyclohexanone, Acetic acid [(lR,2S,5R)-2-isopropyl-5-methylcyclohexyl] ester, l,3,3-Trimethyl-2- oxabicyclo[2,2,2]octane, and (R)-l-methyl-4-(l-methylethenyl)cyclohexene. Excipient 4 in specific compositions is H-(0-CH2-CH2)n-0H where n is an integer from 3 to 900 (e.g., PEG having an average molecular weight in the range of 2000-12000). These compositions can be filled into soft gel or hard gel capsules depending on its flowability at the temperatures useful for making these dosage forms. These compositions may include a hydrophilic additive.

Shown below are various compositions suitable for oral administration as described herein. In these Examples the amount of excipient adds up to 100% (does not include the API) and the API weight percent is the final weight percent in the pharmaceutical composition.

The API in these compositions in specific compositions is testosterone tridecanoate, testosterone undecanoate, or a combination thereof. Alternatively, the skilled artisan understands that any other testosterone ester or steroid, or steroid ester can be used in these compositions accordingly. Excipient 1 in specific compositions is (9Z)-octadec-9-enoic acid. Excipient 2 in specific compositions is a combination of mono-, di-, or tri- propane-1, 2, 3-triol esters of octadecanoic acid and hexadecanoic acid, octadecanoic acid or a combination thereof. Excipient 3 in specific compositions is H-(0-CH2-CH2)n-0H where n is an integer from 3 to 900 (e.g., PEG having an average molecular weight in the range of 2000-12000) . Excipient 4 in specific compositions is (lR,2S,5R)-2-isopropyl-5-methylcyclohexanol or a combination of one or more of (1R,2S,5R)- 2-isopropyl-5-methylcyclohexanol, (2S,5R)-2-Isopropyl-5-methylcyclohexanone, Acetic acid [(lR,2S,5R)-2-isopropyl-5-methylcyclohexyl] ester, l,3,3-Trimethyl-2-oxabicyclo[2,2,2]octane, and (R)-l-methyl-4-(l-methylethenyl)cyclohexene. These compositions can be filled into soft gel or hard gel capsules depending on its flowability at the temperatures useful for making these dosage forms. Examplary Compositions

Shown below are various compositions suitable for oral administration as described herein. In these Examples the amount of excipient adds up to 100% (does not include the API) and the API weight percent is the final weight percent in the pharmaceutical composition.

The API in these compositions in specific compositions is testosterone tridecanoate, testosterone undecanoate, or a combination thereof. Alternatively, the skilled artisan understands that any other testosterone ester or steroid, or steroid ester can be used in these compositions accordingly. Excipient 1 in specific compositions is (9Z)-octadec-9-enoic acid. Excipient 2 in specific compositions is a combination of mono-, di-, or tri- propane-1, 2, 3-triol esters of octadecanoic acid and hexadecanoic acid, octadecanoic acid, or a combination thereof. Excipient 3 in specific compositions is H-(0-CH2-CH2)n-0H where n is an integer from 3 to 900 (e.g., PEG having an average molecular weight in the range of 2000-12000) . Excipient 4 in specific compositions is (lR,2S,5R)-2-isopropyl-5-methylcyclohexanol or a combination of one or more of (1R,2S,5R)- 2-isopropyl-5-methylcyclohexanol, (2S,5R)-2-Isopropyl-5-methylcyclohexanone, Acetic acid [(lR,2S,5R)-2-isopropyl-5-methylcyclohexyl] ester, l,3,3-Trimethyl-2-oxabicyclo[2,2,2]octane, and (R)-l-methyl-4-(l-methylethenyl)cyclohexene. These compositions can be filled into soft gel or hard gel capsules depending on its flowability at the temperatures useful for making these dosage forms.

Examplaiy Compositions

Shown below are various compositions suitable for oral administration as described herein. In these Examples the amount of excipient adds up to 100% (does not include the API) and the API weight percent is the final weight percent in the pharmaceutical composition

The API in these compositions in specific compositions is testosterone tridecanoate, testosterone undecanoate, or a combination thereof. Alternatively, the skilled artisan understands that any other testosterone ester or steroid, or steroid ester can be used in these compositions accordingly. Excipient 1 in specific compositions is (9Z)-octadec-9-enoic acid, hexadecanoic acid or a combination thereof. Excipient 2 in specific compositions is a combination of mono-, di-, or tripropane- 1,2,3 -triol esters of octadecanoic acid and hexadecanoic acid. Excipient 3 in specific compositions polyoxylated hydrogenated castor oil (Cremophor R40). Excipient 4 in specific compositions is ascorbyl palmitate. These compositions can be filled into soft gel or hard gel capsules depending on its flowability at the temperatures useful for making these dosage forms.

It is to be understood that various formulations of androgen receptor agonists, testosterone and testosterone esters can be used in the methods described herein. Such exemplary formulations can be found e.g., US Patent Application Publication Nos.

20170354663; 20180028542; 20180125787; 20180147215; 20180021349; 20170065614;

20150328233; 20170136033; 20140011780; 20170056415; 20170020893; 20170246187; 20180110786; and 20170106002; each of which is incorporated by reference in its entirety. Examples of such formulations are as follows:

Formulation AAA Ingredients mg/capsule (% w/w): Testosterone Undecanoate 158.3 mg (19.8%); Oleic Acid 413.1 mg (51.6 %); Cremophor RH 40 128.4 mg (16.1%); Borage Seed Oil 80.0 mg (10%); Peppermint Oil 20.0 mg (2.5%); BHT 0.2 mg (0.03%); Total 800 mg (100%).

Formulation AAB Ingredients mg/capsule (% w/w): Testosterone Undecanoate 158.3 mg (19.8%); Oleic Acid 412.5 mg (51.6%); Cremophor RH 40 128.4 mg (16.0%); Peppermint Oil 20.0 mg (2.5%); Borage Seed Oil 80.0 mg (10%); BHT 0.2 mg (0.03%) Ascorbyl Palmitate 0.8 mg (0.1%); Total 800 mg (100%).

Thus, the formulation can comprise: (a) 15-25 percent by weight of a solubilized testosterone undecanoate; (b) 12-18 percent by weight of at least one hydrophilic surfactant; (c) 50-65 percent by weight of at least one lipophilic surfactant; and (d) 10-15 percent by weight of a mixture of borage oil and peppermint oil.

Thus, the formulation can comprise: (a) 18-22 percent by weight of a solubilized testosterone undecanoate; (b) 15-17 percent by weight of at least one hydrophilic surfactant; (c) 50-55 percent by weight of at least one lipophilic surfactant; and (d) 10-15 percent by weight of a mixture of borage oil and peppermint oil.

Thus, the formulation comprises: a) a phytosterol or phytosterol ester; b) a non-sterol solubilizing agent; and c) at least one lipophilic, poorly water soluble therapeutic agent (e.g., a testosterone ester such as testosterone undecanoate or testosterone tridecanaote), in a

composition, wherein the composition is effective to enhance solubility of at least one therapeutic agent, as compared to the solubility of the same therapeutic agent in the absence of a) a phytosterol or phytosterol ester and b) a non-sterol solubilizing agent. Exemplary formulations of type are given in the next two tables.

Some formulation of the formulations described herein include a water soluble (hydrophilic) surfactant, a non-ionic surfactant and a water insoluble (hydrophobic) surfactant. The surfactants can be in any ratio that will give the desired properties of the formulations with the testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate). In certain embodiments of the formulations described herein the ratio by weight of the water soluble surfactant, a non-ionic surfactant and a water insoluble surfactant is about 1:1:1, 1:2:1, 1:3:1, 1:4:1, 1:5:1, 2:1:1, 2:2:1, 2:3:1, 2:4:1, 2:5:1, 3:1:1, 3:2:1, 3:3:1, 3:4:1, 3:5:1, 4:1:1, 4:2:1, 4:3:1, 4:4:1, 4:5:1, 5:1:1, 5:2:1, 5:3:1, 5:4:1 or 5:5:1. In certain embodiments of the formulations described herein the ratio by weight of the water soluble surfactant, a non-ionic surfactant and a water insoluble surfactant is about 1:1:2, 1:2:2, 1:3:2, 1:4:2, 1:5:2, 2:1:2, 2:2:2, 2:3:2, 2:4:2, 2:5:2, 3:1:2, 3:2:2, 3:3:2, 3:4:2, 3:5:2, 4:1:2, 4:2:2, 4:3:2, 4:4:2, 4:5:2, 5:1:2, 5:2:2, 5:3:2, 5:4:2 or 5:5:2. In certain embodiments of the formulations described herein the ratio by weight of the water soluble surfactant, a nonionic surfactant and a water insoluble surfactant is about 1:1:3, 1:2:3, 1:3:3, 1:4:3, 1:5:3, 2:1:3, 2:2:3, 2:3:3, 2:4:3, 2:5:3, 3:1:3, 3:2:3, 3:3:3, 3:4:3, 3:5:3, 4:1:3, 4:2:3, 4:3:3, 4:4:3, 4:5:3, 5:1:3, 5:2:3, 5:3:3, 5:4:3 or 5:5:3. In certain embodiments of the formulations described herein the ratio by weight of the water soluble surfactant, a non-ionic surfactant and a water insoluble surfactant is about 1:1:4, 1:2:4, 1:3:4, 1:4:4, 1:5:4, 2:1:4, 2:2:4, 2:3:4, 2:4:4, 2:5:4, 3:1:4, 3:2:4, 3:3:4, 3:4:4, 3:5:4, 4:1:4, 4:2:4, 4:3:4, 4:4:4, 4:5:4, 5:1:4, 5:2:4, 5:3:4, 5:4:4 or 5:5:4. In certain embodiments of the formulations described herein the ratio by weight of the water soluble surfactant, a nonionic surfactant and a water insoluble surfactant is about 1:1:5, 1:2:5, 1:3:5, 1:4:5, 1:5:5, 2:1:5, 2:2:5, 2:3:5, 2:4:5, 2:5:5, 3:1:5, 3:2:5, 3:3:5, 3:4:5, 3:5:5, 4:1:5, 4:2:5, 4:3:5, 4:4:5, 4:5:5, 5:1:5,

5:2:5, 5:3:5, 5:4:5 or 5:5:5. In certain embodiments the ratio of by weight of the water soluble surfactant, a non-ionic surfactant and a water insoluble surfactant is 1:3:5.

Examples of these types of formulation can have e.g. from 5%-30% and preferably 5%-15% loading (w/w%) of testosterone ester (e.g., testosterone undecanoate or testosterone tridecanoate) and Formulation XA (CAPRYOL 90/TWEEN 80/ TRANSCUTOL P (2:5:5)); Formulation XB (Propylene glycol/CREMOPHOREL/CAPRYOL 90 (1:3:5)); Formulation XC (Sunflower Seed Oil/CREMOPHOR EL/SPAN 80 (3/4/3 w/w/w)); Formulation XD (Com Oil/CREMOPHOR EL/SPAN 80 (3/4/3 w/w/w)); and Formulation E (Sunflower Seed Oil/ CREMOPHOR

EL/Maisine (1:1: 1)). Related formulation are disclosed below (other testosterone esters can be sued including testosterone tridecanoate). Moreover the formulation that do not include testosterone undecanoate below can be adapted to include testosterone esters such as testosterone undecanoate, testosterone tridecanoate or other testosterone esters.

Another composition which can be used with the methods described here include Andriol or Andriol Testocaps which are or were commercially available.

(a) testosterone undecanoate, (b) cholesterol, and (c) at least one phospholipid, wherein (a) and (b) are present in a weight ratio (a):(b) ranging from 1.0.0.05 to 10:0.30 and (a), (b) and

(c) are present in a weight ratio of (a): ((b)+(c)) ranging from 1.0: 1.0 and 1.0:2.5.

In one aspect, the phospholipid is selected from distearoyl phos phatidylcholine, dipalmitoyl phosphatidylcholine, dimyris toyl phosphatidylcholine, egg phosphatidylcholine, soy phosphatidylcholine, dimyrsityl phosphatidyl glycerol Sodium, 1,2-dimyristoyl-phosphatidic acid, dipalmitoyl phosphatidylglycerol, dipalmitoyl phosphate, 1,2-dis tearoyl-sn-glycero-3- phospho-rac-glycerol. 1,2-distearoyl sn-glycero-3-phosphatidic acid, phosphatidylserine and sphingomyelin, or a combination thereof. In one aspect, the phospholipid is distearoyl phosphatidylcholine.

An oral pharmaceutical composition comprising a testosterone derivative having a log P of at least 5, wherein the testosterone derivative is selected from testosterone undecanoate, testosterone enanthate, testosterone oleate, or testosterone palmitate, wherein the testosterone derivative is present in an amount from 0.5% to 20% by weight based on 100% total weight of the composition, and a vehicle, wherein the vehicle comprises (a) a fat component in an amount of at least 500 mg sufficient to achieve lymphatic absorption in a mammal, wherein the fat component comprises a mono-glyceride of long chain fatty acids, a tri-glyceride of long chain fatty acids, or a mono- and triglyceride of long chain fatty acids, wherein the long chain fatty acids in the monoglycerides are selected from fatty acid chains having from 14 to 22 carbon atoms and the long chain fatty acids in the triglycerides are selected from fatty acid chains having from 14 to 22 carbon atoms, and (b) a hydrophilic surfactant, wherein the hydrophilic surfactant is selected from hydrogenated castor oil ethoxylates, polysorbates or any other hydrophilic surfactant with a Hydrophile-Lipophile Balance (HLB) value of 10 or higher, and any combination thereof, wherein the weightratio (a):(b) is from 10: 1 to 1 : 1.

It is also to be understood that although this disclosure is described mainly in the context of oral testosterone/testosterone ester treatments, other routes of administration used in the art can be adapted to the methods described in this disclosure.

The following examples are provided to promote a more clear understanding of certain embodiments of this disclosure and are in no way meant as a limitation thereon. EXAMPLES

Example 1 : Clinical Trial

Oral testosterone therapy was evaluated in a multicenter, phase 3, randomized, open- label, active controlled, parallel-group study that included a 13 -week efficacy phase and a 52- week safety phase. An active arm in which subjects received T gel was included for safety evaluation only.

Materials and Methods

Study Centers and Ethics

Forty US sites enrolled subjects from February 2014 to April 2015. Each site received

Institutional Review Board approval of the protocol before study commencement. Study Population

Males 18 through 80 years old with a documented diagnosis of primary hypogonadism

(congenital or acquired) or hypogonadotropic hypogonadism (congenital or acquired) and with morning serum T<300 ng/dL based on 2 consecutive blood samples obtained between 0600 and 1000 on 2 separate days at approximately the same time were eligible to participate. Subjects were either naive to T therapy or completed an adequate washout of T therapy (eg, 12 weeks after intramuscular androgen injections excluding TU injections which required a longer waiting period; 4 weeks after topical or buccal androgens; and 3 weeks after oral androgens). Exclusion criteria included abnormal prostate digital rectal examination with palpable nodule(s) or

International Prostate Symptom Score (IPSS) >19 points; body mass index >38 kg/m 2 , clinically significant laboratory values (baseline hemoglobin <11.5 or >16.5 g/dL, hematocrit <35% or >54%, serum transaminases >2.5 times upper limit of normal, serum bilirubin >2.0 mg/dL, creatinine >2.0 mg/dL, prostate specific antigen [PSA] >2 ng/mL, prolactin >17.7 ng/mL);

history of seizures or convulsions; any surgical procedure that might interfere with

gastrointestinal motility, pH, or absorption; stroke or myocardial infarction within 5 years;

current or suspected prostate or breast cancer; severe, untreated, obstructive sleep apnea; long QT syndrome or unexplained sudden death in first-degree relative; concurrent medication use that could impact absorption, distribution, metabolism, or excretion of TU or place subject at risk for treatment with T; and dose changes of antihypertensive, lipid-lowering, or hypoglycemic agents within previous 3 months.

Study Design and Procedures Subjects were randomized in a 2: 1 ratio to oral testosterone undecanoate or T gel. oral testosterone undecanoate subjects started at a dose of 225 mg TU (“testosterone undecanoate”) taken twice daily (total daily dose: 450 mg TU) every 12 hours. For optimal lymphatic absorption oral testosterone undecanoate must be administered with a meal [15,16] Therefore, subjects were instructed to administer oral testosterone undecanoate after a standard meal (defined as a meal consisting of 800 to 1400 calories with approximately 20% to 35% fat content). The oral testosterone undecanoate dose was titrated at Weeks 4 and 8, if necessary, based on results of intensive 24-hour PK blood sampling obtained at Weeks 3 and 7,

respectively. Oral testosterone undecanoate subjects stayed at the clinic overnight during each intensive PK blood sampling period. Dose titration was based on serum T average concentration over 24 hours (Cavg0-24h) and maximum observed serum concentration (Cmax) and was titrated by 75 mg dose adjustments. Oral testosterone undecanoate dose levels permitted during the study were 150 mg BID, 225 mg BID, and 300 mg BID. Data collected during an intensive 24-hour PK sampling overnight stay at Week 13 were used for efficacy analysis; dose was not adjusted at this visit. Additional outpatient visits were conducted at Weeks 26, 39, and 52 during which single blood draws were performed 3 to 6 hours post morning dose. Dose was not adjusted during these visits.

T gel (AndroGel ® 1.62%, Abb Vie, Chicago, Illinois, US) was supplied in non-aerosol, metered dose pumps that deliver 20.25 mg T per complete pump actuation. T gel was dosed according to product labeling. Subjects receiving T gel completed outpatient visits at Weeks 2 and 4 with single blood draws to determine titration and at Weeks 3 and 5 for dose changes, if necessary. They also completed an outpatient visit at Week 7 for safety assessments and at Weeks 13, 26, 39 and 52 during which single blood samples were obtained 3 to 6 hours after the morning dose. Dose was not adjusted during these visits. Overnight clinic stays were not required for T gel subjects.

Assessments

Safety endpoints included adverse events (AEs), physical examination findings, vital signs, electrocardiograms (ECGs), and clinical laboratory tests. AEs were classified using the Medical Dictionary for Regulatory Activities (Version 16.1). Safety endpoints were summarized using descriptive statistics. Lab parameters were evaluated using two sample t-tests comparing change from baseline to end of study (EOS) across oral testosterone undecanoate and T gel subjects with p-values <0.05 considered statistically significant.

Hormone Assessment and Statistical Analysis

Serum levels of T, TU, dihydrotestosterone (DHT), dihydrotestosterone undecanoate (DHTU), and estradiol (E2) were evaluated in oral testosterone undecanoate treated subjects.

Pharmacokinetic parameters were calculated for T Cmax and T Cavg0-24h. Sex hormone binding globulin (SHBG) was evaluated in data from all subjects. Serum T concentrations at screening were measured using chemiluminescence immunoassay kits. Hormone assays for pharmacokinetic assessment were analyzed at PPD Bioanalytical Laboratory in Richmond, Virginia. Serum samples were analyzed for T, DHT, TU, DHTU, and E2 using a validated liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay. Subjects

Demographic characteristics were similar for oral testosterone undecanoate and T gel subjects (Table 1). The study was completed by 61.9% and 67.6% of oral testosterone undecanoate treated and T gel subjects, respectively. Non-safety related reasons for early discontinuation included lost to follow-up (6.7% and 11.4%), consent withdrawn due to confinement or schedule conflict (6.7% and 2.9%), consent withdrawn due to reason other than confinement or schedule (4.8% and 1.9%), met Cmax or Cavg stopping criteria (4.3% of oral testosterone undecanoate treated subjects and not applicable for T gel subjects), protocol deviation (3.3% and 2.9%), lack of efficacy (2.4% and 3.8%), and subject enrolled at more than one study site (1.9% and 2.9%). Table 2 presents safety-related reasons for early discontinuation.

Pharmacokinetic Assessments

Efficacy and pharmacokinetic results of this study are described elsewhere [14] In brief, at Week 13, serum T Cavg0-24h was between 300 and 1140 ng/dL in 87.4% of LPCN 1021 subjects (95% lower bound Cl: 81.7%), which met predefined efficacy criteria. Week 13 CavgO- 24h was 446 ng/dL and Cmax was 1134 ng/dL. Average serum T levels remained within eugonadal levels throughout the study (481±371, 543±438, and 538±545 ng/dL at Weeks 26, 39, and 52, respectively). Calculated free T levels, were consistently within normal values, 9 to 30 ng/dL. Measured DHT concentrations increased as T concentrations increased, indicating that T converts to DHT via 5-alpha reductase activity leading to DHT pharmacokinetics that simulated those of T. Overall ratios of DHT/T for oral testosterone undecanoate treated subject based on Cavg0-24h at Weeks 3, 7, and 13 were 0.24, 0.26, and 0.26, respectively, and based on Cmax at Weeks 3, 7, and 13 were 0.16, 0.18, and 0.18, respectively, which were within the normal range for DHT/T ratio of 0.05 to 0.33. Serum E2 concentrations for oral testosterone undecanoate treated subjects remained within normal levels throughout Week 52 for oral testosterone undecanoate treated subjects.

Mean SHBG levels decreased at each post-baseline visit following oral testosterone undecanoate administration, but not T gel (Figure 1). The largest decrease in oral testosterone undecanoate treated subjects occurred at Week 13 (change from baseline of -11.75 nmol/L), after which SHBG levels increased slightly and remained relatively stable. Lipoprotein-associated phospholipase A2 decreased significantly from baseline to EOS for both oral testosterone undecanoate and T gel, but the difference in the change from baseline between groups was not significant.

Table 1 Trial. Demographic and Baseline Characteristics

Table 2 Trial. Mean (SD) Change from Baseline to End of Study in Androgenic Parameters, Liver Enzymes and Lipid Parameters (Safety Set, N = 314)

Mean hematocrit increase from baseline to EOS was similar for treatments. To minimize excessive hematocrit increases, investigators could prematurely withdraw subjects with hematocrit >54%. Elevated hematocrit (>54%) occurred in 8 oral TU treated subjects and 1 T gel subject: 3 of these 8 oral TU treated subjects (1.4%) and the T gel subject (1.0%) were withdrawn; 3 oral TU treated subjects were not withdrawn because hematocrit levels decreased at a subsequent laboratory evaluation; and 2 subjects were withdrawn due to reasons other than elevated hematocrit. Small and similar increases in hemoglobin from pretreatment to EOS occurred in oral TU and T gel subjects.

Serum HDL cholesterol showed a significantly larger decrease at EOS for oral testosterone undecanoate than T gel. For HDL cholesterol, 19.0% of oral testosterone

undecanoate subjects and 14.5% of T gel subjects had a shift from normal at baseline to low at EOS. No statistically significant differences between oral testosterone undecanoate and T gel were found between treatments for other lipid parameters. Oral Testosterone Undecanoate Trial Results - Liver Function Enzymes and Lipids Analysis

Table 3 Trial. Mean change from baseline of liver function enzymes for 4 th quartile patients based on lab parameter level in 1 year oral testosterone undecanoate study

Example 2: Clinical Trial

The following study was designed and performed to evaluate twice-daily dosing of 225 mg TU in a population of male subjects having testosterone deficiency. This trial was conducted in a manner similar to that of Example 1.

All subjects received 225 mg TU BID (two capsules of 112.5 mg) taken twice daily (total daily dose of 450 mg taken as 225 mg in the morning and 225 mg in the evening), approximately 12 hours apart, approximately 30 minutes after morning and evening meals, with water.

Labs were collected at baseline and end of study. Subjects were dosed for 24 days. In the Figure legends, this trial is referred to as“16-002”.

Example 3: Clinical Trial

The following study was designed and performed to evaluate thrice-daily dosing of 150 mg TU (450 mg TU per day) in a population of male subjects having testosterone deficiency. This trial was conducted in a manner similar to that of Example 1. All subjects received 150 mg TU BID (two capsules of 75 mg) taken thrice (total daily dose of 450 mg taken as 150 mg after breakfast, 150 mg after lunch and 150 mg after dinner

approximately 30 minutes after meals, with water.

Labs were collected at baseline and end of study. Subjects were dosed for 24 days. In the Figure legends to this trial is referred to as“16-003”. Example 4: Clinical Trial

This was a multi-center, open-label, randomized, parallel, 2-period, multiple-dose study that evaluated the safety, tolerability, and PK of oral testosterone tridecanoate in 5 groups of hypogonadal males. Period 1 and Period 2 each include an up to 28-day Screening Period, a 14- day Treatment Period, and an Exit Evaluation performed following the final PK blood sample collection.

In Period 1, 3 groups of approximately 12 hypogonadal male subjects each (approximately 36 subjects total) were randomized to receive administration of either Treatment A (500 mg oral testosterone tridecanoate), Treatment B (750 mg oral testosterone tridecanoate), or Treatment C (1000 mg oral testosterone tridecanoate) once daily (QD) for 14 days following standardized meals. Following completion of Period 1, the appropriate dose of oral testosterone tridecanoate for Treatment D (either 250 mg or 1250 mg oral testosterone tridecanoate) in Period 2 was determined based on an evaluation of Period 1 data. Subjects who completed Period 1 proceeded to Period 2 following a 2-day washout.

In Period 2, two groups of approximately 12 hypogonadal male subjects each (approximately 24 subjects total) will be randomized to receive administration of either Treatment D (250 mg or 1250 mg oral testosterone tridecanoate) QD for 14 days following standardized meals, or Treatment E (500 mg oral testosterone tridecanoate) twice daily (BID) for 14 days following standardized meals.

Each subject in Period 1 or Period 2 underwent 3 overnight clinic confinements (on Day 1, Day 8, and Day 14). Subjects entered the clinic at least 2 hours prior to the anticipated morning dosing and remained in confinement for 24 hours after the morning dose. During this time, subjects received each assigned dose of oral testosterone tridecanoate, 30 minutes (plus/minus 5 minutes) after starting a standardized meal. Blood samples for PK analysis were collected at regular intervals for 24 hours after the morning dose. Following the Hour 24 blood draw, subjects were discharged from the clinic. At the end of the Day 1 and Day 8 clinic confinements, subjects received a sufficient quantity of oral testosterone tridecanoate to administer their assigned treatment at home until the next confinement. Subjects also received specific instructions on the standardized meals (approximate composition and/or example meals from various restaurants and instructions for preparation of meals) to be consumed approximately 30 minutes prior to oral testosterone tridecanoate administration, at approximately the same-time of day as the administration during confinement. In addition to the 3 confinements, subjects in Period 1 returned to the clinic on the morning of Day 6, Day 7, Day 12, and Day 13 for a single pre-dose blood sample collection.

The total number of subjects planned for Period 1 of this study is approximately 36;

approximately 24 of these subjects are expected to continue participation into Period 2. If this is not the case, additional subjects are enrolled for Period 2 at the discretion of the Sponsor.

The maximum treatment duration for a subject who participates in both treatment periods will be 28 days (i.e., 14 days for each period). The maximum duration of subject participation depended on the number of days between the end of Period 1 and the beginning of Period 2. This is intended to be less than or equal to 30 days; however, subjects will be informed at the time of consent that this duration may extend beyond 30 days. Assuming 30 days between periods, the maximum duration of subject participation is 86 days (i.e., up to 28 days for screening, 14 days for Period 1, 30 days between periods, and 14 days for Period 2). In the Figure legends this trial is referred to as“1111” or“1111 QD”.

Example 5: Pharmaceutical Composition, Formulations and Unit Dosage Forms The API in this example in specific compositions is testosterone tridecanoate, testosterone undecanoate, or a combination thereof. Alternatively, the skilled artisan understands that any other testosterone ester or steroid, or steroid ester can be used in this compositions accordingly.

These compositions can be made by any suitable method and filled into hard gel or soft gel capsules as appropriate. For example, the one or more of the ingredients are warmed or heated to a temperature that allows for dissolving any solid ingredients, the API is added and mixed until a homogenous mixture is obtained, and the capsule can be filled at an appropriate temperature and if needed, allowed to cool to room temperature.

Composition (A)

Example 7: Pharmaceutical Composition, Formulations and Unit Dosage Forms

The API in this example in specific compositions is testosterone tridecanoate, testosterone undecanoate, or a combination thereof. Alternatively, the skilled artisan understands that any other testosterone ester or steroid, or steroid ester can be used in this compositions accordingly. These compositions can be made by any suitable method and filled into hard gel or soft gel capsules as appropriate. For example, the one or more of the ingredients are warmed or heated to a temperature that allows for dissolving any solid ingredients, the API is added and mixed until a homogenous mixture is obtained and the capsule can be filled at an appropriate temperature and if needed, allowed to cool to room temperature. Composition (F)

Based on the description provided herein and the results of the clinical trial, it is now possible to provide pharmaceutical compositions similar to those described as Compositions (A) - (J) having API The API in this example in specific compositions is testosterone tridecanoate, testosterone undecanoate, or a combination thereof (alternatively, the skilled artisan understands that any other testosterone ester or steroid, or steroid ester can be used in this compositions accordingly) in an amount of e.g., 1 mg to 10 mg, 10 mg to 25 mg, 25 mg to 50 mg, 75 mg to 100 mg, 100 mg to 125 mg, 125 mg to 150 mg, 150 mg to 175 mg, 175 mg to 200 mg, 200 mg to 225 mg, 225 mg to 250 mg, 250 mg to 275 mg, 275 mg to 300 mg, 300 mg to 325 mg, 325 mg to 350 mg, 350 mg to 375 mg, 375 mg to 400 mg, 400 mg to 425 mg, 425 mg to 450 mg, 450 mg to 475 mg, 475 mg to 500 mg, 525 mg to 550 mg, 550 mg to 575 mg, or575 mg to 600 mg.

Other formulation for use in the method include, but are not limited to Formulation XXI Ingredients mg capsule %, w/w Testosterone Undecanoate 158.3 mg (19.8 w/w%) Oleic Acid 413.1 mg (51.6%) Cremophor RH 40 128.4 mg (16.1 w/w%) Borage Seed Oil 80.0 mg (10 w/w%) Peppermint Oil 20.0 mg (2.5%) BHT 0.2 mg (0.03 w/w%) Total 800 mg (100 w/w%) and XX2 Ingredients mg/capsule w/w %, Testosterone Undecanoate 158.3 mg (19.8%) Oleic

Acid 412.5 mg (51.6% w/w) Cremophor RH 40 128.4 mg (16.0 w/w%) Peppermint Oil 20.0 mg (2.5 w/w %) Borage Seed Oil 80.0 mg (10% w/w) 0.03% w/w BHT Ascorbyl Palmitate 0.8 mg (0.1 % w/w) Total 800 mg (100% w/w).

Similar composition to any specific compositions describe herein can also have for example:

(a) a different fatty acid, an additional fatty acid or a both,

(b) a different hydrophilic surfactant, an additional hydrophilic surfactant or both,

(c) a mono- or di-glyceride in place of the fatty acid or in combination with the fatty acid,

(d) a different solidifying agent, an additional solidifying agent, or both,

(e) a different diglyceride than glyceryl palmitostearate, an additional diglyceride or both,

(f) a different antioxidant, an additional antioxidant or both,

(g) have additional additives,

(h) use menthol or another alcohol in place of or in addition to peppermint oil,

(i) use a tocopherol in place of fatty acid, in combination with fatty acid, in place of

peppermint oil, in addition to peppermint oil or a combination thereof, (j) use a different monoglyceride than glyceryl monolinoleate, an additional monoglyceride, a diglyceride in place of glyceryl monolinoleate, a diglyceride in combination with glyceryl monolinoleate or a combination thereof, or

(k) a combination of any of the above.

Moreover, it is noted that any of the formulations disclosed herein can include Vitamin E (e.g., tocopherol), Vitamin E derivatives, Vitamin E prodrugs, Omega-3 fatty acids and the such by adjusting the relative amounts of the other formulation components to achieve the desired formulation.

Other tocopherols, tocotrienols and the such can be sued in this formulations such as d-alpha tocopherol or its acetate as described elsewhere herein.

Example 6: Clinical Study

This example is part of an ongoing Liver Fat Study which is an open-label, multi-center, single arm study evaluating LPCN 1144 treatment (e.g., androgen therapy, testosterone therapy, oral testosterone ester, or 225 mg testosterone undecanoate orally) in a cohort of 36 hypogonadal males (NCT03868059). Subjects with at least 10% baseline liver fat were evaluated which is indicative of subjects with NAFLD with the potential to have NASH. Interim results of seven of the nine subjects in the Liver Fat Study with baseline liver fat of at least 10% are presented as two subjects were unable to schedule an eight-week MRI-PDFF visit. Baseline mean liver fat of these seven subjects was 21.0%.

Baseline Results

An MRI-PDFF study of 36 male hypogonadal subjects (having 2 morning testosterone levels of 300 ng/dL or less) was performed as part of an ongoing trial. Basic baseline labs including liver function tests (e.g., AST & ALT) and serum triglycerides were collected and analyzed. It was found by non-linear regression, that BMI (ALT, AST, serum Triglyceride) was associated with liver fat (P < 0.006). It was found by non-linear regression, that serum triglyceride levels (obesity, hypertension, type 2 diabetes) were associated with liver fat (P < 0.006). The analysis of this data is summarized in the figures.

Interim Results

Treatment results showed an absolute mean reduction from baseline of 7.6% liver fat and demonstrated a 38% relative mean liver fat reduction from baseline. Moreover, there was an 86% responder rate in which subjects experienced at least a 4.1% absolute reduction in liver fat from baseline and a 71% responder rate in which subjects experienced at least a 29% reduction in liver fat from baseline.

End of Study Results

34 subjects were evaluable at end-of-study. 21 subjects had NAFLD at baseline (liver fat greater than or equal to 5%, mean liver fat = 12.1%), 10 subjects had greater than 8% liver fat at baseline (mean = 12.1%), and 8 subjects had greater than 10% liver fat at baseline (mean = 20.5%). Mean relative liver fat % change after 16 weeks of treatment was -33% for subjects having NAFLD (e.g., baseline liver fat of 5% or greater), -42% for baseline liver fat of 8% or greater, and -40% for baseline liver fat of 10% or greater. The responder rates (% of patients with at least a 30% relative reduction in liver fat from baseline) was 71% for greater than or equal to 5% baseline liver fat, 80% for greater than or equal to 8% baseline liver fat, and 70% for greater than or equal to 10% baseline liver fat. NAFLD was resolved (baseline liver fat greater than or equal to 5% going to less than 5% at end of study) in 48% of subject. 100% of subjects with above normal ALT levels (mean baseline = 48.7) ended the study with normal ALT levels. %0% of subjects with above normal GGT levels ended the study with normal GGT levels. 60% of subjects had BUN normalization. The normalization rates for total cholesterol, LDL-C, triglyceride were 29%, 50% and 21% respectively. The mean relative liver fat change is patients with NAFLD resolution was -55%. As the duration of therapy increased from 8 weeks to 16 weeks, liver fat reductions were improved as well as the percent of subject achieving NAFLD resolution.

A recent publication in Therapeutic Advances in Gastroenterology (Patel et al.,

Therapeutic Advances in Gastroenterol. 2016 Sep; 9(5): 692-701) quantified the magnitude of MRI-PDFF reduction corresponding to a histologic response in NASH in a clinical setting with paired liver biopsy. The results concluded that histologic responders had a statistically significant reduction in MRI-PDFF of -4.1% with a mean relative percent change of -29.3%.

This is a particularly surprising and unexpected finding since oral testosterone undecanoate lowers serum SHBG levels - the skilled artisan would expect that lower SHBG would be expected to result in greater liver fat according the art. Thus, the skilled artisan can apply these teaching and examples to employ androgen therapy for treating or preventing liver disease as described herein.

It is understood that the above-described various types of compositions, dosage forms and/or modes of applications are only illustrative of preferred embodiments of the present invention. Numerous modifications and alternative arrangements may be devised by those skilled in the art without departing from the spirit and scope of the present invention and the appended claims are intended to cover such modifications and arrangements. Thus, while the present invention has been described above with particularity and detail in connection with what is presently deemed to be the most practical and preferred embodiments of the invention, it will be apparent to those of ordinary skill in the art that variations including, but not limited to, variations in size, materials, shape, form, function and manner of operation, assembly and use may be made without departing from the principles and concepts set forth herein.