Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS OF USE OF GAMMA-KETOALDEHYDE SCAVENGERS FOR TREATING, PREVENTING OR IMPROVING NONALCOHOLIC FATTY LIVER DISEASE (NAFLD), NASH, ALD OR CONDITIONS RELATED TO THE LIVER
Document Type and Number:
WIPO Patent Application WO/2018/048932
Kind Code:
A1
Abstract:
Methods and compositions for use in treating, preventing or improving diseases related to the liver in an animal, including but not limited to nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD) or nonalcoholic steatohepatitis (NASH), are described. The compounds of the present invention are gamma-ketoaldehyde scavengers.

Inventors:
RATHMACHER JOHN (US)
ABUMRAD NAJI (US)
FLYNN CHARLES (US)
Application Number:
PCT/US2017/050317
Publication Date:
March 15, 2018
Filing Date:
September 06, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
METABOLIC TECH INC (US)
International Classes:
C07C217/58; C07C215/50
Domestic Patent References:
WO2008070778A22008-06-12
Foreign References:
US20150265584A12015-09-24
US6794398B12004-09-21
US20140256774A12014-09-11
US20120157501A12012-06-21
Other References:
ROYCHOWDHURY ET AL.: "Formation of r-ketoaldehyde-protein adducts during ethanol-induced liver injury in mice", FREE RADICAL BIOLOGY AND MEDICINE, vol. 47, no. 11, 17 July 2009 (2009-07-17), pages 1526 - 1538, XP026742368
Attorney, Agent or Firm:
CORYELL, Matthew (US)
Download PDF:
Claims:
Claiming:

1. A method for treating, preventing, or ameliorating liver disease to an animal with or at risk of liver disease, thereby inhibiting or treating the liver disease, comprising the step of administering to the animal and effective amount of the compound having a structure represented by a compound of the following formula:

wherein: R is N or C;

R2 is independently H, hydroxy, halogen, nitro, CF3, C1-6 alkyl, C1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R2, R3 and R4, and may cyclize with to one or more R2, R3, or R5 to form an optionally substituted C3-8 membered ring containing C, O, S or N;

R3 is H, hydroxy, halogen, nitro, CF3, C1-6 alkyl, C1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R4, R2 and R3 may cyclize with to one or more R2 or R5 to form an optionally substituted C3-8 membered ring containing C, O, S or N;

R4 is H, hydroxy, halogen, nitro, CF3, C1-6 alkyl, C1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R4, R2 and R3 may cyclize with to one or more R2, R3, or R5 to form an optionally substituted C3-s membered ring containing C, 0, S or N;

R5 is a bond, H, hydroxy, halogen, nitro, CF3, C1-6 alkyl, C1-6 alkoxy, C3-10 cycloalkyl, C3-s membered ring containing C, O, S or N, optionally substituted with one or more R4, R2 and R3 may cyclize with to one or more R2, R3, or R4 to form an optionally substituted C3-s membered ring containing C, O, S or N; and stereoisomers and analogs thereof.

2. The method of claim 1, wherein the liver disease further comprises nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD) or nonalcoholic steatohepatitis

(NASH).

3. The method of claim 1, wherein the compound is selected from the formula:

wherein:

R is N or C;

R2 is independently H, substituted or unsubstituted alkyl;

R3 is H, halogen, alkoxy, hydroxyl, nitro; R4 is H, substituted or unsubstituted alkyl, carboxyl; and pharmaceutically acceptable salts thereof.

4. The method of claim 1, wherein the compound is salicylamine (2-hydroxybenzylamine or 2-HOBA).

5. The method of claim 1, wherein the compound is selected from the formula:

or a pharmaceutically acceptable salt thereof.

6. The method of claim 1, wherein the compound is selected from the formula:

or a pharmaceutically acceptable salt thereof.

7. The method of claim 1, wherein the compound is selected from the formula:

or a pharmaceutically acceptable salt thereof.

8. The method of claim 1, wherein the compound is selected from the formula:

or a pharmaceutically acceptable salt thereof.

9. The method of claim 1, wherein the compound is selected from the formula:

Salicylamine Methylsalicylamine

5- Methoxysalicyl 3- Methoxysalicyl (SA) (MeSA)

(5-MoSA) (3-MoSA)

Ethylsalicylamine Pyridoxamine Ethylpyridoxamine Pentylpyridoxa (EtSA) (PM) (EtPM) (PPM)

or a pharmaceutically acceptable salt thereof.

10. A nutritional composition comprising a pharmaceutically acceptable carrier and a

compound of the following formula, and its use as agents in a method for treating,

preventing, or ameliorating liver disease to an animal with or at risk of liver disease,

thereby inhibiting or treating the liver disease:

wherein: R is N or C; R2 is independently H, hydroxy, halogen, nitro, CF3, C1-6 alkyl, C1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R2, R3 and R4, and may cyclize with to one or more R2, R3, or R5 to form an optionally substituted C3-8 membered ring containing C, O, S or N;

R3 is H, hydroxy, halogen, nitro, CF3, C1-6 alkyl, C1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R4, R2 and R3 may cyclize with to one or more R2 or R5 to form an optionally substituted C3-8 membered ring containing C, O, S or N;

R4 is H, hydroxy, halogen, nitro, CF3, C1-6 alkyl, C1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R4, R2 and R3 may cyclize with to one or more R2, R3, or R5 to form an optionally substituted C3- 8 membered ring containing C, O, S or N;

R5 is a bond, H, hydroxy, halogen, nitro, CF3, C1-6 alkyl, C1-6 alkoxy, C3-10 cycloalkyl, C3- 8 membered ring containing C, O, S or N, optionally substituted with one or more R4, R2 and R3 may cyclize with to one or more R2, R3, or R4 to form an optionally substituted C3- 8 membered ring containing C, O, S or N; and stereoisomers and analogs thereof.

Description:
COMPOSITIONS AND METHODS OF USE OF GAMMA-KETOALDEHYDE SCAVENGERS FOR TREATING, PREVENTING OR IMPROVING NONALCOHOLIC FATTY LIVER DISEASE (NAFLD), NASH, ALD OR CONDITIONS RELATED TO

THE LIVER

This application claims priority to U.S. Application Serial No. 62/383,895 filed

September 6, 2016 and to U.S. Application Serial No. 62/410,133 filed October 19, 2016 which are herein incorporated by reference in their entireties.

Background of the Invention

1. Field

The present invention relates to a composition comprising a γ-ketoaldehyde (γ-ΚΑ) scavenging compound, such as 2-Hydroxybenzylamine (2-HOBA), and methods of

administering a γ-ΚΑ scavenger to treat, prevent or improve diseases or conditions relating to the liver including nonalcoholic fatty liver disease (NAFLD) and/or alcoholic liver disease (ALD), and/or nonalcoholic steatohepatits (NASH).

2. Background

Chronic liver disease due to alcohol is a leading cause of morbidity and mortality that continues to rise. Second to viral hepatitis, chronic ethanol overconsumption is responsible for 25% of all deaths caused by liver cancer and cirrhosis. Chronic alcohol consumption is a frequent comorbidity of liver disease and cancer. The burden of health care for alcoholic liver disease (ALD) is high with cost estimates approaching $27 billion per year in the U.S. alone. Abstinence is the best therapy for ALD but recidivism is a major risk with relapse rates ranging from 67%-81% over the course of a year.

ALD includes a range of hepatic manifestations including fatty liver (steatosis), hepatitis and cirrhosis/fibrosis that may present simultaneously in a given individual. The spectrum of ALD ranges from simple steotosis to alcoholic steatohepatitis (ASH) to cirrhosis and is aggravated with obesity. There are many mechanisms by which alcohol induces liver injury; however, inflammation underpins the advancement of ALD. Ethanol metabolism promotes antioxidant depletion and leads to the formation of injurious entities including acetaldehyde, acetate, reactive oxygen species (ROS), and lipid peroxides that induce inflammatory responses. Additionally, alcohol and its metabolites incite inflammation by promoting gut leakiness and stimulating immune cells (the so-called adaptive immune response) and/or activating innate immune pathways, such as complement. While activation of innate immunity components initiates alcoholic liver injury it also triggers hepatoprotective, regenerative, and antiinflammatory responses that reduce hepatocyte damage.

It is well known that lipid peroxidation and oxidative stress play significant roles in inflammation and the pathogenesis of chronic liver disease, especially ALD. Aldehydes such as malondialdehyde (MDA) and 4-hydroxynonenol (4-HNE) form covalent protein adducts which interfere with normal protein function. Orders of magnitude more reactive than MDA and 4- HNE are the peroxidation products of arachidonic acid, termed acyclic γ-KAs (also known as isolevuglandins or isoketals), which are key mediators of inflammation (Fig. 1). γ-KAs adduct rapidly and covalently to proteins and DNA, interfere with normal molecule function, and form protein-protein cross-links (isoketals). γ-KAs are produced by the F 2 -Isoprostane (F 2 -IsoP) pathway. The γ-KAs have been shown to accumulate in various pathophysiological conditions through the non-classic eicosanoids, isoprostanes (IsoP) and isofurans (IsoF) that are formed non-enzymatic ally by free radical mediated peroxidation of arachidonic acid. Isofurans are similar to the isoprostanes, but contain a substituted tetrahydrofuran ring. It has been

demonstrated that anti-γ-ΚΑ antibody titers in the serum of human subjects with ALD are elevated relative to subjects without ALD (Fig. 2).

ALD is characterized by the development of steatosis, inflammation, hepatocyte necrosis and apoptosis, with the eventual development of fibrosis and cirrhosis. It is also well established that consumption of alcohol in excess causes an oxidative injury to the liver. F 2 -IsoPs have been shown to be the most accurate predictors of oxidative stress in vivo, and their levels are increased in alcoholic liver disease, and chronic hepatitis. Over-production of KAs is implicated in the pathogenesis of several chronic inflammatory diseases. More recently, ethanol feeding in the mouse has been shown to induce formation of hepatic γ-KAs which readily bind to proteins to form stable adducts. These γ-ΚΑ -protein adducts are likely to contribute to ethanol-induced liver injury by eliciting proinflammatory responses or adduct- specific immune responses.

There is considerable interest in identifying appropriate therapeutic interventions aimed at inhibiting the inflammatory processes and interrupting the immunogenic pathways associated with ALD. 2-hydroxy-benzylamine (2-HOBA), a staple of buckwheat, was found to be a potent scavenger of γ-KAs scavenging γ-KAs 980-fold faster than the rate of formation of γ-ΚΑ-lysyl- protein adducts. Importantly, they showed that this γ-ΚΑ scavenger does not inhibit

cyclooxygenase enzymes. In a model of oxidant mediated cell death (Fig. 3), 2-HOBA almost completely prevented cell death induced by t-butylhydroperoxide (tBHP). In addition, it was demonstrated that 2-HOBA has a protective effect against oxidant mediated cell death HepG2 cells exposed to varying concentrations of hydrogen peroxide (H 2 0 2 ). Despite the profound economic and health impacts of ALD, little progress has been made in the management of patients with this severe clinical condition. While abstinence is a cornerstone of treatment, there is considerable interest in identifying other therapeutic interventions and treatments for ALD. Current therapeutic modalities for ALD include corticosteroids and pentoxyfilline. Corticosteroids improve short-term survival of severe forms of alcoholic hepatitis, but are frequently contraindicated. Pentoxyfilline, a competitive nonselective phosphodiesterase inhibitor, improved short-term survival in severe acute alcoholic hepatitis and demonstrated improved risk-benefit profiles compared to prednisone, but when combined with prednisone it did not confer additional benefit. Both of these treatments attenuate the inflammatory response but do not target the underlying inflammatory signal(s). The trapping of toxic oxidized lipids by 2-HOBA is novel in that it attenuates the formation of aggravating protein adducts that sustain inflammation and drive liver injury. The present invention includes use of 2-HOBA for preventing ALD and also attenuating the propagation of alcoholic liver disease.

Epidemiological data indicate that nonalcoholic fatty liver disease (NAFLD) is the most prevalent form of chronic liver disease in western countries. The spectrum of NAFLD ranges from simple steatosis to nonalcoholic steatohepatits (NASH) to cirrhosis and occurs frequently in the setting of obesity, dyslipidemia and insulin resistance. NASH can lead to cirrhosis and liver failure in 10-15% of patients. The mechanisms discriminating steatosis from NASH are still not entirely understood. It is increasingly evident that inflammation and the consequent production of reactive oxygen species (ROS) and reactive lipid species (RLS) are important components in the pathogenesis of NASH. In the liver, the changes in the inflammatory and immune responses exacerbate ROS and pro-inflammatory cytokine production leading to worsening of NASH. Further, patients with NAFLD and NASH have higher mortality and morbidity in comparison to the general population; NAFLD has increased cardiovascular mortality, NASH has more liver-related mortality. Recent observations concluded that NASH, currently the third most common indication for liver transplantation in the United States, is projected to become the most common indication for liver transplantation in the next 10 years. NASH is the

inflammatory form of NAFLD and is characterized by excess liver fat, inflammation, and hepatocellular ballooning with or without fibrosis. NASH is most concerning for progression to end stage liver disease, or cirrhosis. The mechanisms and conditions favoring NASH are unclear but histologically, it bears resemblance to alcoholic steatohepatitis. It is well-known that lipid peroxidation and oxidative stress play significant roles in the pathogenesis of chronic liver disease including NASH. Reactive oxygen species (ROS) accelerate the formation of lipid peroxides, leading to generation of bifunctional electrophiles (BFEs) that are key mediators of inflammation. Among these BFEs, 4-hydroxynonenal (4-HNE), acrolein, malondialdehyde (MDA), methylglyoxal (MGO) and levuglandins (LGs) are known to mediate oxidative injury by covalently modifying lipids, proteins and DNA. BFEs are extremely reactive compounds that adduct covalently to proteins and DNA, interfere with normal molecule function, and form protein-, phosphoethanolamine- and DNA-cross-links.

Nonalcoholic steatohepatitis (NASH) is liver inflammation and damage caused by a buildup of fat in the liver. NASH resembles ALD but occurs in people who consume little or no alcohol. NASH affects two to five percent of Americans, most often in people who are middle- aged and overweight or obese. The present invention includes use of 2-HOBA for preventing and/or treating NASH. The present invention includes use of 2-HOBA to scavenge toxic oxidized lipids (ketoaldehydes) to effectively regulate the inflammatory program and lead to reversal in the associated hepatic injury.

The present invention includes use of 2-HOBA to target γ-ΚΑ to prevent lipid peroxidation and the resulting γ-ΚΑ-specific immune responses in alcoholic liver disease.

Brief Description of the Figures

Figure 1 is a graphic depicting how γ-KAs react with lysine or other primary amines to form a reversible Schiff base adducts.

Figure 2 is a graph depicting serum γ-ΚΑ antibody titers in hospitalized ICU patients with and without alcoholic liver disease.

Figure 3 is a graph depicting the protective effect of 2-HOBA against oxidant mediated cell death in mouse hepatocytes increasing t-butylhydroperoxide.

Figure 4A-B depicts ROS is HepaRG with increasing ethanol dose and the protective effect of 2-HOBA with increasing ethanol concentration.

Figure 5A-B depicts ALT and AST levels in the media of HepaRG cells pretreated with different levels of 2-HOBA.

Figure 6 shows the effects of 2-HOBA pretreatment against ethanol-mediated γ-ΚΑ formation.

Figure 7 shows the identification of γ-ΚΑ-modified histone-H3 and-H4 in mouse lung.

Figure 8 is a graph depicting liver and kidney isoprostanes (IsoP) and isofurans (IsoF). Figure 9 depicts evidence for IsoLG formation in human NAFLD livers.

Figure 10 depicts increased MPO-positive cells in NASH. Figure 11 A-I depicts the development of NAFLD in STAM mice.

Figure 12A-T describes the effects of 2-HOBA on STAM mice.

Figure 139A-F shows insulin signaling and hepatic inflammasomes in STAM

Figure 14A-G depicts the development of NAFLD in DIAMOND™ mice.

Figure 15 summarizes results of DIAMOND™ mice testing. Figure 16 summarizes results of DIAMOND™ mice testing.

Detailed Description of the Invention

The present invention includes a novel nutritional therapy that will reduce liver injury by preventing the formation of γ-ΚΑ-protein adducts and differential effects on innate and adaptive immune responses. This nutritional therapy can be used to treat, prevent or improve conditions or diseases relating to the liver including but not limited to NAFLD, ALD and NASH. The nutritional therapy can be used to improve overall liver health and support healthy liver function.

The present invention comprises a means to specifically prevent the formation of γ-ΚΑ - adducts in the liver using a class of bifunctional electrophile (BFE) "scavenger" molecules. A series of phenolic amines that includes pyridoxamine and its water soluble derivative 2- hydroxybenzylamine (2-HOBA), a natural product of buckwheat seed comprise the preferred embodiment. 2-HOBA in particular reacts 980-fold faster with γ-KAs than with lysine, preventing protein and lipid adduction in vitro and in vivo. The compositions and methods of this invention are directed to animals, including human and non-human animals. The animal may be healthy or may be suffering from a disease or condition.

The term administering or administration includes providing a composition to a mammal, consuming the composition and combinations thereof.

The present invention includes compositions and methods of use of 2-HOBA, alternatively named salicylamine, SAM, 2-hydroxylbenzylamine, and pentylpyridoxamine (PPM).

Embodiments of the present invention include compounds of the following formula, and their use as agents in a method for treating, preventing, or ameliorating liver conditions or diseases including NAFLD, ALD and NASH to a subject with or at risk of liver conditions or diseases including NAFLD, ALD and NASH, thereby inhibiting or treating the liver conditions or diseases:

wherein:

R is N or C;

R 2 is independently H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C 3 -s membered ring containing C, O, S or N, optionally substituted with one or more R 2 , R 3 and R 4 , and may cyclize with to one or more R 2 , R 3 , or R5 to form an optionally substituted C 3 -8 membered ring containing C, O, S or N;

R 3 is H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R 4 , R 2 and R 3 may cyclize with to one or more R 2 or R5 to form an optionally substituted C3-8 membered ring containing C, O, S or N;

R 4 is H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R 4 , R 2 and R 3 may cyclize with to one or more R 2 , R 3 , or R5 to form an optionally substituted C3-8 membered ring containing C, O, S or N;

R5 is a bond, H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C3- 8 membered ring containing C, O, S or N, optionally substituted with one or more R 4 , R 2 and R3 may cyclize with to one or more R 2 , R3, or R 4 to form an optionally substituted C3-8 membered ring containing C, O, S or N; and stereoisomers and analogs thereof.

Another embodiment of the present invention includes compounds of the following formula, and their use in methods for treating, preventing, or ameliorating liver conditions or diseases including NAFLD, ALD and NASH to a subject with or at risk of these liver conditions:

wherein:

R is N or C;

R 2 is independently H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R 2 , R 3 and R 4 , and may cyclize with to one or more R 2 , R 3 , or R5 to form an optionally substituted C 3 -8 membered ring containing C, O, S or N;

R 3 is H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R 4 , R 2 and R 3 may cyclize with to one or more R 2 or R5 to form an optionally substituted C3-8 membered ring containing C, O, S or N;

R 4 is H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C3-8 membered ring containing C, O, S or N, optionally substituted with one or more R 4 , R 2 and R3 may cyclize with to one or more R 2 , R3, or R5 to form an optionally substituted C3-8 membered ring containing C, O, S or N;

R5 is a bond, H, hydroxy, halogen, nitro, CF3, C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C3- 8 membered ring containing C, O, S or N, optionally substituted with one or more R 4 , R 2 and R3 may cyclize with to one or more R 2 , R 3 , or R 4 to form an optionally substituted C 3 -s membered ring containing C, O, S or N; and stereoisomers and analogs thereof.

In certain embodiments, the compound may be selected from the compounds disclosed herein. In a preferred embodiment, the compound may be salicylamine.

Another embodiment of the present invention is a method for treating, preventing, or ameliorating liver conditions or diseases including NAFLD, ALD and NASH to a subject with or at risk of liver conditions or diseases including NAFLD, ALD and NASH, thereby inhibiting or treating the liver conditions, comprising the step of co-administering to the subject at least one compound in a dosage and amount effective to treat the dysfunction in the mammal, the compound having a structure represented by a compound of the following formula:

wherein:

R is N or C;

R 2 is independently H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C 3 -s membered ring containing C, O, S or N, optionally substituted with one or more R 2 , R 3 and R 4 , and may cyclize with to one or more R 2 , R 3 , or R5 to form an optionally substituted C 3 -8 membered ring containing C, O, S or N; R 3 is H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C 3 -s membered ring containing C, O, S or N, optionally substituted with one or more R 4 , R2 and R 3 may cyclize with to one or more R 2 or R5 to form an optionally substituted C 3 -s membered ring containing C, O, S or N;

R 4 is H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C 3 -s membered ring containing C, O, S or N, optionally substituted with one or more R 4 , R 2 and R 3 may cyclize with to one or more R 2 , R 3 , or R5 to form an optionally substituted C 3 -s membered ring containing C, O, S or N;

R5 is a bond, H, hydroxy, halogen, nitro, CF 3 , C 1-6 alkyl, C 1-6 alkoxy, C3-10 cycloalkyl, C3- 8 membered ring containing C, O, S or N, optionally substituted with one or more R 4 , R 2 and R 3 may cyclize with to one or more R 2 , R 3 , or R 4 to form an optionally substituted C3-8 membered ring containing C, O, S or N; and stereoisomers and analogs thereof; with a drug having a known side effect of treating, preventing, or ameliorating liver conditions or diseases including NAFLD, ALD and NASH.

Examples of compounds that may be used with the methods disclosed herein include, but are not limited to, compounds selected from the formula:

wherein: R is N or C;

R 2 is independently H, substituted or unsubstituted alkyl; R 3 is H, halogen, alkoxy, hydroxyl, nitro;

R 4 is H, substituted or unsubstituted alkyl, carboxyl; and pharmaceutically acceptable salts thereof.

In a preferred embodiment, the compound is salicylamine (2-hydroxybenzylamine or 2- HOBA).

The compound may be chosen from:

or a pharmaceutically acceptable salt thereof.

The compound may also be chosen from:

or a pharmaceutically acceptable salt thereof.

The compounds or analogs may also be chosen from:

pharmaceutically acceptable salt thereof.

The compounds may also be chosen from:

pharmaceutically acceptable salt thereof. The compounds may also be chosen from

Salicylamine Methylsalicylamine

5- Methoxysalicylamine 3- Methoxysalicyl (SA) (MeSA)

(5-MoSA) (3-MoSA)

Ethylsalicylamine Pyridoxamine Ethylpyridoxamine Pentylpyridoxa (EtSA) (PM) (EtPM) (PPM)

or a pharmaceutically acceptable salt thereof.

The compounds of the present invention can be administered by any method and such methods are well known to those skilled in the art and include, but are not limited to oral administration, transdermal administration, administration by inhalation, nasal administration, topical administration, intravaginal administration, ophthalmic administration, intraaural administration, intracerebral administration, rectal administration, and parenteral administration, including injectable administration such as intravenous administration, intra-arterial

administration, intramuscular administration and subcutaneous administration. The compounds can be administered therapeutically, to treat an existing disease or condition, or prophylactically for the prevention of a disease or condition. Although any suitable pharmaceutical medium comprising the composition can be utilized within the context of the present invention, preferably, the composition is combined with a suitable pharmaceutical carrier, such as dextrose or sucrose.

Methods of calculating the frequency by which the composition is administered are well- known in the art and any suitable frequency of administration can be used within the context of the present invention (e.g., one 6 g dose per day or two 3 g doses per day) and over any suitable time period (e.g., a single dose can be administered over a five minute time period or over a one hour time period, or, alternatively, multiple doses can be administered over an extended time period). The composition of the present invention can be administered over an extended period of time, such as weeks, months or years. The composition can be administered in individual servings comprising one or more than one doses (individual servings) per day, to make a daily serving comprising the total amount of the composition administered in a day or 24 hour period.

Any suitable dose of the present composition can be used within the context of the present invention. Methods of calculating proper doses are well known in the art.

"Treatment" or "treating" refers to the medical management of a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder. This term includes active treatment, that is, treatment directed specifically toward the improvement of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder.

Experimental Examples

Example 1

Fully-differentiated, 21 -day, HepaRG hepatocytes (Bio-Predict International) display clear epithelial cells surrounded by hepatocyte colonies that contain numerous bile canaliculi which readily metabolize the fluorescent MRP2 substrate, CDFDA. HepaRG cells are a unique and well-established human hepatic cell culture system with high fidelity to the absorption, distribution, metabolism, and excretion of human liver. Consistent with the known mechanisms of action of 2-HOB A, a 24 hr pre-treatment of HepaRG cells with 2-HOBA (0, 250, 500 and 1000 μΜ) was followed by increasing ethanol concentration (0-200 mM; 24 hr) but did not affect reactive oxygen species formation (Fig. 4A). However, 2-HOBA almost completely prevented cell death by ethanol (Fig. 4B). Similarly, a 24 hr pre-treatment of HepaRG cells with 2-HOBA (0, 250, 500 and 1000 μΜ) followed by increasing ethanol concentration (0-200 mM; 24 hr) resulted in a near normalization of ALT (Fig. 5A) and AST (Fig. 5B) levels in culture media taken from HepaRG hepatocytes. Currently, there is little knowledge concerning the identity of proteins that are susceptible to γ-KAs modification. The present inventors

investigated the identity of proteins adducted endogenously using cultured HepaRG hepatocytes, exposed to ethanol with or without prior 2-HOBA pretreatment (0, 250 or 500 μΜ for 24 hr.). Cells were harvested and total protein was resolved by SDS-PAGE then probed with a well- characterized single chain antibody, Dl l SCFv, which recognizes peptides and proteins modified by γ-KAs isomers (Fig. 6). Consistent with the theory that there is a 'pool' of γ-ΚΑ-modification susceptible proteins, all treatments exhibited a similar level of "background" anti-Dl 1 cross- reactivity; however, there was a robust increase in the cross -reactivity of a single band (arrow) at -18 kD in hepatocytes treated with ethanol but without 2-HOBA protection. In contrast, the intensity of this band was restored to levels observed in untreated controls with 2-HOBA pre- treatment 2-HOBA (250 or 500 μΜ). Interestingly, this band is of the same molecular weight as that recently identified as γ-ΚΑ adducted and anti-Dl 1 antibody in a mouse model of lung fibrosis (Fig. 7). These data signify that ETOH promotes the formation of γ-ΚΑ-adducted proteins, the formation of which can be blocked with 2-HOBA.

Taken together, these findings demonstrate that γ-KAs are major mediators of liver injury caused by lipid peroxidation, and that the use of 2-HOBA protects against cell death induced by such oxidants or ethanol exposure. Targeting γ-KAs may be used as a method for preventing lipid peroxidation and the resulting γ-ΚΑ-specific immune responses in alcoholic liver disease.

Example 2

2-HOBA and ethanol were concurrently administered to C57/BL6J mice to establish the efficacy of 2-HOBA in mitigating ethanol-mediated liver dysfunction (increased AST and ALT), reducing γ-ΚΑ formation, favorably altering immune responses and stimulating beneficial intracellular signaling pathways. The results indicate that pre-treatment with 2-HOBA (1.0 mg/ml) significantly reduces liver injury (elevated γ-KAs and liver function enzymes) activated upon ethanol exposure.

2-HOBA (0.5 g/L in Lieber-DeCarli liquid diet, LDLD) was administered to mice prior to ethanol treatment for 14 days to examine 2-HOBA efficacy in mitigating liver injury. Ethanol (5% v/v) was administered using the NIAAA model (10 days ethanol in LDLD + binge). Food intake and body weight were not significantly impacted with the addition of 0.5 g/L of 2-HOBA to LDLD (data not shown). White blood cell and lymphocyte cell counts were reduced while eosinophil and basophil counts were increased with ethanol relative to maltodextrin feeding (Table 1), consistent with other reports of alcoholic liver injury. Such responses were not observed in the presence of 2-HOBA.2-HOBA at 0.5 g/L did not attenuate increases in serum AST or ALT in these studies (Table 2).

Table 1. Clinical blood chemistry of mice after the NIAAA ethanol treatment regimen.

Con + MD Con + ETOH 2-HOBA + MD 2-HOBA + ETOH

WBC 5.70 ±0.61 2.47 ± 0.33** 6.08 ±1.05 3.11 ±0.66

Lymphocytes 65.67 ±2.63 50.18 ±2.98** 58.14 ±5.45 50.42 ± 4.59

Monocytes 7.97 ±0.65 8.29 ±1.32 6.66 ±0.92 6.33 ±1.11

Eosinophils 0.96 ±0.16 3.61 ±0.69*** 1.79 ±0.40 2.71 ±0.46

Basophils 0.40 ±0.10 1.74 ±0.66* 0.60 ±0.14 1.27 ±0.26*

Abbreviations: Cell counts are in (χ10 3 /μΙ). WBC; white blood count; Statistical significance was determined using a two-tailed unpaired Student's t-test. N= 12 mice per group. P≤ 0.01 vs Con + MD;

***P≤ 0.001 vs Con + MD; # P≤ 0.05 vs 2-HOBA+MD.

Table 2. Primary endpoints in mice after the NIAAA treatment regimen with or without 2-HOBA (0.5 g/L).

(1) Con + MD (2) Con + ETOH (3) 2-HOBA + MD (4) 2-HOBA + 1 vs2 3 vs4 1 vs3 2 vs4

ETOH

AST 992.8 ±345.1 961.1 ±297.7 566.5 ±34.1 1725.0 ±300.2 0.93 0.0017"* 0.34 0.98

ALT 77.2 ±15.1 406.4 ±77.2 54.3 ±20.1 443.7 ± 66.2 0.0018" 0.0001" 0.29 0.75

*

Data reported as mean ± S.E.M. AST, serum aspartate aminotransferase; ALT, alanine aminotransferase; N= 12 mice per group. ** P indicates significance≤0.01, *** P indicates significance≤ 0.001 by unpaired Student's two- tailed t-test. AST and ALT are in U/L.

An additional cohort of mice were administered a 14-day pretreatment of 2-HOBA (1.0 g/L) prior to administration of ethanol using the NIAAA model. Mice administered ethanol consumed less food and consequently weighed ~ 2.5 g less than maltodextrin-fed controls at the end of study.

Table 3. Primary endpoints in mice the NIAAA treatment regimen with or without 2-HOBA (1.0 g/L).

(1)Con + MD (2)Con+ (3) 2-HOBA + MD (4) 2-HOBA + 1 vs2 3vs4 1 vs3 2vs4

ETOH ETOH

AST 634.4 ± 95.0 906.1 ±186.1 693.6 ± 173.1 676.6 +86.7 0.17 0.94 0.76 0.30 ALT 50.2 ±5.5 253.9 ±62.9 127.5 ±78.59 164.4 ±20.8 0.0014"* 0.72 0.35 0.21 BUN 21.3 ±1.2 27.1 ±5.5 34.6 ±2.9 22.6 ±1.7 0.25 0.60 0.30 0.47 CRE 0.4 ± 0.01 0.2 ± 0.05 0.3 ± 0.05 0.3 ± 0.04 0.0003" * 0.67 0.19 0.06

Data reported as mean ±S.E.M. AST, serum aspartate aminotransferase; ALT, alanine aminotransferase; N= 12 mice per group. *** P indicates significance≤ 0.001 by unpaired Student's two-tailed t-test. AST and ALT concentrations are in U/L. Blood urea nitrogen (BUN) and creatinine (CRE) units are mg/dL.

Hepatic triglyceride content trended to be lower in ETOH-challenged mice pre-treated with 2-HOBA (1.0 g/L)). Liver damage as indicated by changes in serum ALT trended lower with 2-HOBA pre-treatment (164.4 ± 20.8 U/L) versus Con + ETOH mice vs (253.0 ± 62.9 U/L) but these did not reach statistical significance. AST was lower in mice pretreated with 2-HOBA + ETOH (676.6 ±86.7 U/L) compared to controls (906.1 ± 186.1 U/L) but these did not reach statistical significance. These changes were accompanied with significant reductions in hepatic isoprostanes (IsoP) and isofurans (IsoF) and also resulted in significant reductions in kidney isoprostane levels (1.3 ± 0.1 ng/g versus 2.0 ± 0.3 ng/g in Con + ETOH; <0.05) as shown in Fig. 8.

Pre-treatment with 2-HOBA for 14 days at 1.0 g/L in the NIAAA model reduced liver injury and ameliorated the significant increases in hepatic isoprostane and isofuran content observed with ETOH exposure and additionally decreased kidney isoprostane formation.

Example 3-NASH

F2-Isoprostanes (F2-IsoPs) are prostaglandin-like compounds formed in vivo via a non- enzymatic mechanism involving the free radical-initiated peroxidation of arachidonic acid and have been shown to be the most accurate predictors of oxidative stress in vivo. Elevated levels of plasma or urinary F2-IsoPs have been reported in alcoholic liver disease patients and in animals with NAFLD/ NASH and increased liver F 2 -IsoP also have been reported in humans and animals with NASH. F 2 -IsoPs values are also increased in chronic hepatitis. Preliminary data show that liver obtained from mice and human liver tissue with proven NAFLD/NASH show dramatically increased levels of IsoLG protein adducts relative to controls (Fig. 9). These observations were concurrent with increased myeloperoxidase-positive neutrophil staining in specimens that is hallmark of biopsy-proven NASH (Fig. 10), suggestive of enhanced inflammation. Several agents appear to be associated with this enhanced inflammatory process.

It has been demonstrated that increased plasma lipopoly saccharide (LPS) and Free Fatty Acids (FFAs) in plasma of NASH patients which were associated with increased hepatic TLR4- Myd88-independent signaling. It has also demonstrated in cultured HepaRG cells that palmitate and LPS induce NF-kB activity that can be blocked with chemical- or small-interfering RNA- mediated inhibition of TLR4. Liang et al recently demonstrated that alternative metabolic ligands (cholesterol and carbohydrate) also may trigger enhanced steatosis, hepatocellular hypertrophy, and mixed-type (neutrophilis and mononuclear cell) inflammation through an inflammasome-mediated mechanism.

Inflammasomes are multimeric protein complexes of the innate immune system that upon PAMP (pathogen-associated molecular pattern) or DAMP (damage-associated molecular pattern) binding, either directly or through the adaptor molecule ASC, activating caspase 1. Caspase 1 in turn activates downstream signaling pathways and the resultant changes in biological function vary considerably depending upon cell type and initiating ligand. Inflammasomes are activated by members of the (NOD)-like receptor (NLR) family (NLRPl-3, NLRP6-7, NLRP12, NLRC4), NAIP and Aim2. Activation of the NLRP3 inflammasome leads to caspase- 1 cleavage of inactive pro-IL-Ιβ, pro-IL-18 and pro-IL-33 into their active forms. Fatty acids, cholesterol, and protein aggregates are among the known NLRP3 -inflammasome activators. The described molecular triggers of NASH inflammasomes include DNA, saturated fatty acids and LPS, NLRP-1. This suggests that IsoLGs are also additional potent inflammasome activators through mechanisms that are not fully delineated. Indeed, defective NLRP1- and NLRP3- signaling/activation by IsoLG ligands may underpin altered liver immunometabolism, host gut microbiome adaptations, and defective NLRP-inflammasome sensing that are hallmark of the NAFL to NASH transition.

: 2-Hydroxybenzylamine (2-HOBA) is 980 times more reactive than lysine with yKAs and importantly, does not inhibit cyclooxygenase enzymes. In a model of oxidant mediated cell death 2-HOBA almost completely prevented hepatocyte cell death induced by t- butylhydroperoxide (tBHP). This is a remarkable finding given the fact the pathogenesis of oxidative injury is quite complex and multifaceted. In addition, it was demonstrated that 2- HOBA has a protective effect against oxidant mediated cell death in HepG2 cells exposed to varying concentrations of H2O2. Taken together, these findings suggest that yKAs are potentially major mediators of liver injury caused by lipid peroxidation, and that the use of 2-HOBA would protect against cell death induced by such oxidants in the liver. Obviously, the efficacy of 2- HOBA depends on its accumulation in target tissues, and this was recently supported by pharmacokinetic studies in an in vivo mouse model. Administration of 2-HOBA at 1, 3, and 10 g/L for 7 days was associated with dose-dependent increases in plasma and liver levels of 2- HOBA. The levels in the liver were about 10 times higher than in plasma.

One model used in studying the pathogenesis of NASH, cirrhosis and HCC is the STAM model, (Stelic Inc. Tokyo, Japan). With strong fidelity to human NASH both histologically and physiologically (elevated fasting blood sugar, liver biochemistries, intrahepatic lipid, dyslipidemia; Figure 11), the STAM model develops the features previously difficult to obtain in genetic knockouts or dietary models of NAFLD. The present inventors have demonstrated that 2-HOBA can reduce NAFLD severity in STAM mice (Figure 12).

In the STAM™ model, NASH is induced in C57BL/6 mice by a single subcutaneous injection of 200 μg streptozotocin (STX) solution 2 days after birth and feeding with high-fat diet beginning at 4 wks of age. At 3 wks of age, 12 mice that had undergone STZ injection were divided into two groups: 1) 2-HOBA (n=6), and 2) vehicle control (n=6). Mice in the 2-HOBA group received 2-HOBA in drinking water (lg/L water), while the vehicle control group received plain water without 2-HOBA. At 4 wks of age, all mice were placed on ad libitum high fat diet. All mice remained on the high fat diet and received 2-HOBA-supplemented or plain water based on their group assignment throughout the study protocol. Body weights and food/water intake were monitored weekly. Animals were sacrificed at 9 wks of age (6 weeks of 2-HOBA or vehicle treatment), and tissues and serum were collected for analysis.

Liver sections were stained with hematoxylin and eosin (for scoring of steatosis, hepatocyte ballooning, and inflammation), Sirius red (for assessment of fibrosis), and for F4/80+ macrophages. Scoring was performed in a blinded manner for steatosis, ballooning, inflammation, and necrosis using the following criteria, Steatosis (0-4): 0 = <5%; 1 = 5-25%; 2 = 25-50%; 3 = 50-75%; 4 = 75-100%. Ballooning (0-3): 0 = absent; 1 = mild (focal involving fewer than three hepatocytes); 2 = moderate (focal involving more than three hepatocytes or multifocal); 3 = prominent (multifocal with more than two foci of three or more hepatocytes). Inflammation (0-4): 0 = absent; 1 = minimal (zero to one focus per 20x field); 2 = mild (two foci); 3 = moderate (three foci); 4 = severe (four or more foci). Serum levels of glucose, insulin, alanine transaminase, aspartate transaminase, triglycerides, cholesterol, and F2-isoprostanes were measured. Serum and tissue levels of the following inflammatory markers were measured by multiplex assay (Luminex, Millipore, Billerica, MA): IL-la, IL-6, IL-Ιβ, IL- 10, IL- 17, MCP-1, and TNFa. Liver 2-HOBA, F 2 -isoprostane, and isofuran levels were determined by LC/MS/MS methods. Liver mRNA expression was assessed via RT-qPCR for the following genes: Gck, Pckl, Pdk4, Irsl , Irs2, Pgcla, Cptla, Gyk, Srepblc, Accl , Fxr, Coxl, Cox2, Nox4, Catalase, Gpxl, Gp41phx, and p22phox. Liver protein content and phosphorylation status were determined for Akt, GSK3P, mTOR, ERK, JNK, FOXOl, and NALP. Two-tailed independent samples t-tests were used to compare endpoints between 2-HOBA and vehicle treated groups. Significance was set at a = 0.05.

The viability, clinical signs and behavior were monitored daily. Mean body weights and food intake were similar between groups, however liver weight, and liver to body weight ratios were significantly reduced (Figure 12). Serum glucose levels were similar between groups, but insulin, ALT and lipids all were trending towards reduction. Importantly, serum isoprostanes (a marker of BFE formation and IsoLG adduction) and histologic NASH severity were significantly reduced with 2-HOBA treatment.

The livers from STAM mice were subjected to immunoblot analysis to better understand the basis for the improvements in liver function and phenotypes described in Fig. 13. 2-HOBA treatment dramatically increased AKT and GSK3P phosphorylation without significantly altering signaling through mTOR, ERK or FOXOl . In addition, the data show significantly decreased expression of pyruvate dehydrogenase kinase 4 (pdk4), a key protein in the regulation of mitochondrial fuel metabolism, suggesting that the reductions in liver weight, improvements in NASH severity and reductions in serum isoprostanes with 2-HOBA treatment may be mediated by an γΚΑ-mediated mechanism affecting mitochondria function. Furthermore, the data show a decrease NALP1 with 2HOBA treatment. A role for NRLP3-inflammasomes in regulating mitochondrial function was recently described. These data in the STAM model signify that IsoLG-protein adducts cause injury to the liver, and that 2-HOBA potently reduces NASH severity and restores markers of hepatic insulin sensitivity. Importantly, the data show NLRPl-inflammasome activation in this model is blocked with oral 2-HOBA administration suggesting a role for 2-HOBA in blocking IsoLG-mediated inflammasome NASH responses.

Example 4

DIAMOND (Diet Induced Animal Model of Non-alcoholic fatty liver Disease) is a proprietary isogenic mouse strain that sequentially develops non-alcoholic fatty liver disease, nonalcoholic steatohepatitis, fibrosis, and hepatocellular carcinoma in response to a high-fat, high- sugar diet. Disease progression in the DIAMOND mice uniquely parallels human disease progression, including histopathology. The DIAMOND mouse model is unique in that it is the only murine model of NAFLD/NASH that develops NASH solely as a result of the Western diet (high fat, sugar water) with no gene knockouts or toxins to induce liver pathology (Fig. 14).

Twelve 8-wk old male DIAMOND mice were placed on ad libitum high fat diet (Harlan - ENVIGO TD.88317) and water containing glucose (18.9% w/v) and fructose (23.1% w/v); all mice remained on this diet throughout the study protocol. At 12 wks of age, mice were divided into two groups: 1) 2-HOBA (n=6), and 2) vehicle controls (n=6). Animals in the 2-HOBA group received 2-HOBA in drinking water (lg/L water with glucose and fructose). The vehicle control group received water without 2-HOBA (with glucose and fructose). Body weight and food intake were measured weekly. At -23 wks of age, all animals underwent a glucose tolerance test (GTT) and MRI imaging to assess hepatic fat. For the GTT, animals were fasted for 12 hours and then glucose (2 g/kg bw of a 100 mg/mL glucose in sterile water) was administered by oral gavage. Blood was sampled at 0, 15,30, 45, 60, 90, and 120 minutes after glucose administration and area under the curve was calculated. Animals were sacrificed at 24 wks of age (12 weeks of 2-HOBA or vehicle treatment). Tissues and serum were collected for analysis.

Liver sections were stained with hematoxylin and eosin (for scoring of steatosis, hepatocyte ballooning, and inflammation) and Sirius red (for assessment of fibrosis). Scoring was performed in a blinded manner for steatosis, ballooning, inflammation, and necrosis using the following criteria, Steatosis (0-4): 0 = <5%; 1 = 5-25%; 2 = 25-50%; 3 = 50-75%; 4 = 75-100%. Ballooning (0-3): 0 = absent; 1 = mild (focal involving fewer than three hepatocytes); 2 = moderate (focal involving more than three hepatocytes or multifocal); 3 = prominent (multifocal with more than two foci of three or more hepatocytes). Inflammation (0-4): 0 = absent; 1 = minimal (zero to one focus per 20x field); 2 = mild (two foci); 3 = moderate (three foci); 4 = severe (four or more foci). Serum levels of glucose, alanine transaminase, and aspartate transaminase were measured. Liver mRNA expression was assessed via RT-qPCR for the following genes: Tnfa, Nlrpla, Mb, 1118, Timpl, Collal, ProCard, Nlrp3, Caspl, Prolllb, Tgfbl, Bambi, Pdk4, and Gapdh. Two-tailed independent samples t-tests were used to compare endpoints between 2-HOBA and vehicle treated groups. Significance was set at a = 0.05.

Figures 15-16 summarizes the results of the DIAMOND mice testing. These data demonstrate a trend for a reduction in liver weight which was accompanied a significant reduction in the liver enzymes ALT and AST with 2HOBA supplementation. These findings support the efficacy of 2-HOBA to prevent the development or attenuate the severity of NASH. Reference List

Albano E, Vidali M. Immune mechanisms in alcoholic liver disease. Genes Nutr 2010; 5(2):141- 147. PM : 19809845

Mottaran E, Stewart SF, Rolla R et al. Lipid peroxidation contributes to immune reactions associated with alcoholic liver disease. Free Radic Biol Med 2002; 32(1):38-45.PM:11755315

Stewart SF, Vidali M, Day CP, Albano E, Jones DE. Oxidative stress as a trigger for cellular immune

responses in patients with alcoholic liver disease. Hepatology 2004; 39(1):197-203.PM :14752838

Lee GS, Yan JS, Ng RK, Kakar S, Maher JJ. Polyunsaturated fat in the methionine-choline-deficient diet influences hepatic inflammation but not hepatocellular injury. J Lipid Res 2007; 48(8):1885- 1896.PM:17526933

1. Williams R. The pervading influence of alcoholic liver disease in hepatology. Alcohol Alcohol 2008;

43(4):393-397.PM:18385413

2. Murray CJ, Richards MA, Newton JN et al. UK health performance: findings of the Global Burden of Disease Study 2010. Lancet 2013; 381(9871):997-1020.PM:23668584

3. Basra S, Anand BS. Definition, epidemiology and magnitude of alcoholic hepatitis. World J Hepatol 2011; 3(5):108-113.PM:21731902

4. Mackie J, Groves K, Hoyle A et al. Orthotopic liver transplantation for alcoholic liver disease: a retrospective analysis of survival, recidivism, and risk factors predisposing to recidivism. Liver Transpl 2001; 7(5):418-427.PM:11349262

5. Miguet M, Monnet E, Vanlemmens C et al. Predictive factors of alcohol relapse after orthotopic liver transplantation for alcoholic liver disease. Gastroenterol Clin Biol 2004; 28(10 Pt 1):845- 851.PM :15523219

6. Miller WR, Walters ST, Bennett M E. How effective is alcoholism treatment in the United States? J Stud Alcohol 2001; 62(2):211-220.PM:11327187

7. O'Shea RS, Dasarathy S, McCullough AJ. Alcoholic liver disease. Hepatology 2010; 51(1):307- 328.PM :20034030

8. Wang HJ, Gao B, Zakhari S, Nagy LE. Inflammation in alcoholic liver disease. Annu Rev Nutr 2012;

32:343-368. PM:22524187

9. Roychowdhury S, McMullen MR, Pritchard MT et al. An early complement-dependent and TLR-4- independent phase in the pathogenesis of ethanol-induced liver injury in mice. Hepatology 2009; 49(4):1326-1334.PM :19133650

10. Cohen Jl, Roychowdhury S, McMullen MR, Stavitsky AB, Nagy LE. Complement and alcoholic liver disease: role of Clq in the pathogenesis of ethanol-induced liver injury in mice. Gastroenterology 2010; 139(2):664-74, 674.PM:20416309

11. Pritchard MT, McMullen MR, Medof ME, Stavitsky A, Nagy LE. Role of complement in ethanol- induced liver injury. Adv Exp Med Biol 2008; 632:175-186.PM :19025122

12. Gao B, Bataller R. Alcoholic liver disease: pathogenesis and new therapeutic targets.

Gastroenterology 2011; 141(5):1572-1585.PM:21920463

13. Lieber CS. Alcoholic fatty liver: its pathogenesis and mechanism of progression to inflammation and fibrosis. Alcohol 2004; 34(1):9-19.PM:15670660 Rouach H, Fataccioli V, Gentil M, French SW, Morimoto M, Nordmann R. Effect of chronic ethanol feeding on lipid peroxidation and protein oxidation in relation to liver pathology. Hepatology 1997; 25(2):351-355.PM :9021946

Seki S, Kitada T, Yamada T, Sakaguchi H, Nakatani K, Wakasa K. In situ detection of lipid peroxidation and oxidative DNA damage in non-alcoholic fatty liver diseases. J Hepatol 2002; 37(1):56-62.PM :12076862

Benedetti A, Comporti M, Esterbauer H. Identification of 4-hydroxynonenal as a cytotoxic product originating from the peroxidation of liver microsomal lipids. Biochim Biophys Acta 1980;

620(2):281-296.PM:6254573

Janero DR. Malondialdehyde and thiobarbituric acid-reactivity as diagnostic indices of lipid peroxidation and peroxidative tissue injury. Free Radic Biol Med 1990; 9(6):515-540.PM:2079232 Morrow JD, Awad JA, Boss HJ, Blair IA, Roberts LJ. Non-cyclooxygenase-derived prostanoids (F2- isoprostanes) are formed in situ on phospholipids. Proc Natl Acad Sci U S A 1992; 89(22):10721- 10725.PM:1438268

Roychowdhury S, McMullen MR, Pritchard MT, Li W, Salomon RG, Nagy LE. Formation of gamma- ketoaldehyde-protein adducts during ethanol-induced liver injury in mice. Free Radic Biol Med 2009; 47(11):1526-1538.PM:19616618

Morrow JD, Hill KE, Burk RF, Nammour TM, Badr KF, Roberts U. A series of prostaglandin F2-like compounds are produced in vivo in humans by a non-cyclooxygenase, free radical-catalyzed mechanism. Proc Natl Acad Sci U S A 1990; 87(23):9383-9387.PM:2123555

Bertola A, Mathews S, Ki SH, Wang H, Gao B. Mouse model of chronic and binge ethanol feeding (the NIAAA model). Nat Protoc 2013; 8(3):627-637.PM :23449255

Fessel JP, Hulette C, Powell S, Roberts LJ, Zhang J. Isofurans, but not F2-isoprostanes, are increased in the substantia nigra of patients with Parkinson's disease and with dementia with Lewy body disease. J Neurochem 2003; 85(3):645-650.PM:12694390

Binder CJ. Naturally occurring IgM antibodies to oxidation-specific epitopes. Adv Exp Med Biol 2012; 750:2-13. PM:22903662

Chou MY, Hartvigsen K, Hansen LF et al. Oxidation-specific epitopes are important targets of innate immunity. J Intern Med 2008; 263(5):479-488.PM:18410591

Lieber CS. Role of oxidative stress and antioxidant therapy in alcoholic and nonalcoholic liver diseases. Adv Pharmacol 1997; 38:601-628. PM:8895826

Wu D, Cederbaum Al. Alcohol, oxidative stress, and free radical damage. Alcohol Res Health 2003; 27(4):277-284.PM:15540798

Milne GL, Sanchez SC, Musiek ES, Morrow JD. Quantification of F2-isoprostanes as a biomarker of oxidative stress. Nat Protoc 2007; 2(1):221-226.PM:17401357

Ivester P, Roberts LJ, Young T et al. Ethanol self-administration and alterations in the livers of the cynomolgus monkey, Macaca fascicularis. Alcohol Clin Exp Res 2007; 31(1):144-155.PM :17207113 Raszeja-Wyszomirska J, Safranow K, Milkiewicz M, Milkiewicz P, Szynkowska A, Stachowska E. Lipidic last breath of life in patients with alcoholic liver disease. Prostaglandins Other Lipid Mediat 2012; 99(1-2):51-56.PM:22706383

Konishi M, Iwasa M, Araki J et al. Increased lipid peroxidation in patients with non-alcoholic fatty liver disease and chronic hepatitis C as measured by the plasma level of 8-isoprostane. J

Gastroenterol Hepatol 2006; 21(12):1821-1825.PM :17074020

Davies SS, Bodine C, Matafonova E et al. Treatment with a gamma-ketoaldehyde scavenger prevents working memory deficits in hApoE4 mice. J Alzheimers Dis 2011; 27(1):49- 59.PM:21709376

Salomon RG. Isolevuglandins, oxidatively truncated phospholipids, and atherosclerosis. Ann N Y Acad Sci 2005; 1043:327-342. PM:16037255 Salomon RG, Subbanagounder G, O'Neil J et al. Levuglandin E2-protein adducts in human plasma and vasculature. Chem Res Toxicol 1997; 10(5):536-545.PM :9168251

Smathers RL, Galligan JJ, Shearn CT et al. Susceptibility of L-FABP-/- mice to oxidative stress in early-stage alcoholic liver. J Lipid Res 2013; 54(5):1335-1345.PM:23359610

Stewart SF, Vidali M, Day CP, Albano E, Jones DE. Oxidative stress as a trigger for cellular immune responses in patients with alcoholic liver disease. Hepatology 2004; 39(1):197-203.PM :14752838 Mottaran E, Stewart SF, Rolla R et al. Lipid peroxidation contributes to immune reactions associated with alcoholic liver disease. Free Radic Biol Med 2002; 32(1):38-45.PM :11755315 Chedid A, Mendenhall CL, Moritz TE et al. Cell-mediated hepatic injury in alcoholic liver disease. Veterans Affairs Cooperative Study Group 275. Gastroenterology 1993; 105(1):254- 266.PM :8514042

Lin F, Taylor NJ, Su H et al. Alcohol dehydrogenase-specific T-cell responses are associated with alcohol consumption in patients with alcohol-related cirrhosis. Hepatology 2013; 58(1):314- 324.PM :23424168

Rao R. Endotoxemia and gut barrier dysfunction in alcoholic liver disease. Hepatology 2009; 50(2):638-644.PM:19575462

Hritz I, Mandrekar P, Velayudham A et al. The critical role of toll-like receptor (TLR) 4 in alcoholic liver disease is independent of the common TLR adapter MyD88. Hepatology 2008; 48(4):1224- 1231.PM:18792393

Soares JB, Pimentel-Nunes P, Roncon-Albuquerque R, Leite-Moreira A. The role of

lipopolysaccharide/toll-like receptor 4 signaling in chronic liver diseases. Hepatol Int 2010; 4(4):659-672.PM:21286336

Wright SD, Ramos RA, Tobias PS, Ulevitch RJ, Mathison JC. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 1990; 249(4975):1431- 1433.PM:1698311

Markiewski MM, Lambris JD. The role of complement in inflammatory diseases from behind the scenes into the spotlight. Am J Pathol 2007; 171(3):715-727.PM:17640961

Li W, Laird JM, Lu L et al. Isolevuglandins covalently modify phosphatidylethanolamines in vivo: detection and quantitative analysis of hydroxylactam adducts. Free Radic Biol Med 2009;

47(11):1539-1552.PM:19751823

Bykov I, Jauhiainen M, Olkkonen VM et al. Hepatic gene expression and lipid parameters in complement C3(-/-) mice that do not develop ethanol-induced steatosis. J Hepatol 2007;

46(5):907-914.PM:17321001

Bykov I, Junnikkala S, Pekna M, Lindros KO, Meri S. Complement C3 contributes to ethanol- induced liver steatosis in mice. Ann Med 2006; 38(4):280-286.PM :16754259

Kalant D, Cain SA, Maslowska M, Sniderman AD, Cianflone K, Monk PN. The chemoattractant receptor-like protein C5L2 binds the C3a des-Arg77/acylation-stimulating protein. J Biol Chem 2003; 278(13):11123-11129.PM:12540846

Schieferdecker HL, Schlaf G, Koleva M, Gotze O, Jungermann K. Induction of functional anaphylatoxin C5a receptors on hepatocytes by in vivo treatment of rats with IL-6. J Immunol 2000; 164(10):5453-5458.PM :10799912

Mack C, Jungermann K, Gotze O, Schieferdecker HL. Anaphylatoxin C5a actions in rat liver: synergistic enhancement by C5a of lipopolysaccharide-dependent alpha(2)-macroglobulin gene expression in hepatocytes via IL-6 release from Kupffer cells. J Immunol 2001; 167(7):3972- 3979.PM:11564816

Zagol-lkapitte I, Amarnath V, Jadhav S, Oates JA, Boutaud O. Determination of 3- methoxysalicylamine levels in mouse plasma and tissue by liquid chromatography-tandem mass spectrometry: application to in vivo pharmacokinetics studies. J Chromatogr B Analyt Technol Biomed Life Sci 2011; 879(15-16):1098-1104.PM:21489890

Davies SS, Brantley EJ, Voziyan PA et al. Pyridoxamine analogues scavenge lipid-derived gamma- ketoaldehydes and protect against H202-mediated cytotoxicity. Biochemistry 2006; 45(51):15756- 15767.PM:17176098

Carithers L, Jr., Herlong HF, Diehl AM et al. Methylprednisolone therapy in patients with severe alcoholic hepatitis. A randomized multicenter trial. Ann Intern Med 1989; 110(9):685- 690.PM :2648927

Lucey MR, Mathurin P, Morgan TR. Alcoholic hepatitis. N Engl J Med 2009; 360(26):2758- 2769.PM:19553649

Mathurin P, Mendenhall CL, Carithers RL, Jr. et al. Corticosteroids improve short-term survival in patients with severe alcoholic hepatitis (AH): individual data analysis of the last three randomized placebo controlled double blind trials of corticosteroids in severe AH. J Hepatol 2002; 36(4):480- 487.PM :11943418

Naveau S, Chollet-Martin S, Dharancy S et al. A double-blind randomized controlled trial of infliximab associated with prednisolone in acute alcoholic hepatitis. Hepatology 2004; 39(5):1390- 1397.PM:15122768

Mathurin P, Abdelnour M, Ramond MJ et al. Early change in bilirubin levels is an important prognostic factor in severe alcoholic hepatitis treated with prednisolone. Hepatology 2003;

38(6):1363-1369.PM :14647046

Akriviadis E, Botla R, Briggs W, Han S, Reynolds T, Shakil O. Pentoxifylline improves short-term survival in severe acute alcoholic hepatitis: a double-blind, placebo-controlled trial.

Gastroenterology 2000; 119(6):1637-1648.PM:11113085

De BK, Gangopadhyay S, Dutta D, Baksi SD, Pani A, Ghosh P. Pentoxifylline versus prednisolone for severe alcoholic hepatitis: a randomized controlled trial. World J Gastroenterol 2009; 15(13):1613- 1619.PM:19340904

Mathurin P, Louvet A, Duhamel A et al. Prednisolone with vs without pentoxifylline and survival of patients with severe alcoholic hepatitis: a randomized clinical trial. JAMA 2013; 310(10):1033- 1041.PM:24026598

Albano E, Vidali M. Immune mechanisms in alcoholic liver disease. Genes Nutr 2010; 5(2):141- 147.PM :19809845

Stewart SF, Vidali M, Day CP, Albano E, Jones DE. Oxidative stress as a trigger for cellular immune responses in patients with alcoholic liver disease. Hepatology 2004; 39(1):197-203.PM :14752838 Stewart SF, Vidali M, Day CP, Albano E, Jones DE. Oxidative stress as a trigger for cellular immune responses in patients with alcoholic liver disease. Hepatology 2004; 39(1):197-203.PM :14752838 Cresci GA, Bush K, Nagy LE. Tributyrin supplementation protects mice from acute ethanol-induced gut injury. Alcohol Clin Exp Res 2014; 38(6):1489-1501.PM:24890666

Lieber CS. The discovery of the microsomal ethanol oxidizing system and its physiologic and pathologic role. Drug Metab Rev 2004; 36(3-4):511-529.PM:15554233

Perrot N, Nalpas B, Yang CS, Beaune PH. Modulation of cytochrome P450 isozymes in human liver, by ethanol and drug intake. Eur J Clin Invest 1989; 19(6):549-555.PM:2515975

Lieberman MA, Marks AD. Ethanol Metabolism. Basic Medical Biochemistry: A Clinical Approach. Wolters Kluwer; 2012.

Russell TD, Schaack J, Orlicky DJ et al. Adipophilin regulates maturation of cytoplasmic lipid droplets and alveolae in differentiating mammary glands. J Cell Sci 2011; 124(Pt 19):3247- 3253.PM:21878492 Zagol-lkapitte IA, Matafonova E, Amarnath V et al. Determination of the Pharmacokinetics and Oral Bioavailability of Salicylamine, a Potent gamma-Ketoaldehyde Scavenger, by LC/MS/MS. Pharmaceutics 2010; 2(1):18-29.PM:21822464

Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J 2008; 22(3):659-661.PM:17942826

Guidance for Industry: Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult Healthy Volunteers. Center for Drug Evaluation and Research. Rockville, MD, USA: U.S. Food and Drug Administration; 2005.