WO2005039598A1 - Method of treating alcoholic liver disease - Google Patents

Method of treating alcoholic liver disease Download PDF

Info

Publication number
WO2005039598A1
WO2005039598A1 PCT/US2004/035139 US2004035139W WO2005039598A1 WO 2005039598 A1 WO2005039598 A1 WO 2005039598A1 US 2004035139 W US2004035139 W US 2004035139W WO 2005039598 A1 WO2005039598 A1 WO 2005039598A1
Authority
WO
WIPO (PCT)
Prior art keywords
pirfenidone
ifn
patient
dosage
effective amount
Prior art date
Application number
PCT/US2004/035139
Other languages
French (fr)
Inventor
Lawrence M. Blatt
Original Assignee
Intermune, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Intermune, Inc. filed Critical Intermune, Inc.
Publication of WO2005039598A1 publication Critical patent/WO2005039598A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha

Definitions

  • the present invention is in the field of alcoholic liver disease, and use of pirfenidone or a pirfenidone analog to treat alcoholic liver disease.
  • Alcoholic liver disease is a major cause of illness and death, and is the most common liver disease in the United States. It is the fourth leading cause of death in the United States, and results in between 20,000 and 40,000 deaths per year. Women are generally more susceptible to alcohol-induced liver damage than men and develop alcoholic liver disease at a more rapid rate having imbibed less alcohol.
  • ALD involves an acute or chronic inflammation of the liver induced by alcohol abuse.
  • ALD is characterized by fatty liver (steatosis), hepatitis, liver fibrosis, and cirrhosis.
  • Alcoholic hepatitis is characterized histologically by hepatocellular necrosis, alcoholic Mallory's hyaline bodies, and an inflammatory reaction with infiltration by polymorphonuclear leukocytes and lymphocytes.
  • the clinical presentation of alcoholic hepatitis varies with the severity of the disease. Common symptoms are weakness, anorexia, weight loss, nausea, vomiting, and diarrhea. Patients often present with fever, jaundice, and tender hepatomegaly.
  • the present invention provides a method of treating alcoholic hepatitis, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in monofherapy or in combination therapy.
  • the invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in combination therapy with a second therapeutic agent other than IFN- ⁇ .
  • the invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog, a Type II interferon receptor agonist, and a side effect management agent (e.g., a palliative agent, or an agent for the treatment, reduction, or avoidance of a side effect caused by any therapeutic agent).
  • a side effect management agent e.g., a palliative agent, or an agent for the treatment, reduction, or avoidance of a side effect caused by any therapeutic agent.
  • the invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of a TNF- antagonist, which therapy further includes administration of an effective amount of a Type II interferon receptor agonist, and/or pirfenidone or a pirfenidone analog.
  • the terms "individual,” “host,” “subject,” and “patient” are used interchangeably herein, and refer to a mammal, including, but not limited to, primates, including simians and humans.
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse affect attributable to the disease.
  • Treatment covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease.
  • alcoholic hepatitis refers to an acute or chronic inflammatory lesion of the liver that occurs in the context of chronic alcohol abuse.
  • alcoholic hepatic fibrosis used interchangeably herein with “alcoholic liver fibrosis,” refers to the growth of scar tissue in the liver that can occur in the context of chronic alcohol abuse.
  • liver function refers to a normal function of the liver, including, but not limited to, a synthetic function, including, but not limited to, synthesis of proteins such as serum proteins (e.g., albumin, clotting factors, alkaline phosphatase, aminotransferases (e.g., alanine transaminase, aspartate transaminase), 5'-nucleosidase, ⁇ - glutaminyltranspeptidase, etc.), synthesis of bilirubin, synthesis of cholesterol, and synthesis of bile acids; a liver metabolic function, including, but not limited to, carbohydrate metabolism, amino acid and ammonia metabolism, hormone metabolism, and lipid metabolism; detoxification of exogenous drugs; a hemodynamic function, including splanchnic and portal hemodynamics; and the like.
  • the term "dosing event" as used herein refers to administration of a therapeutic agent to a patient
  • Continuous delivery as used herein (e.g. , in the context of “continuous delivery of a substance to a tissue”) is meant to refer to movement of drug to a delivery site, e.g., into a tissue in a fashion that provides for delivery of a desired amount of substance into the tissue over a selected period of time, where about the same quantity of drug is received by the patient each minute during the selected period of time.
  • Controlled release as used herein (e.g., in the context of “controlled drug release”) is meant to encompass release of substance (e.g., a Type II interferon receptor agonist, e.g., IFN- ⁇ ) at a selected or otherwise controllable rate, interval, and/or amount, which is not substantially influenced by the environment of use.
  • substance e.g., a Type II interferon receptor agonist, e.g., IFN- ⁇
  • Controlled release thus encompasses, but is not necessarily limited to, substantially continuous delivery, and patterned delivery (e.g., intermittent delivery over a period of time that is interrupted by regular or irregular time intervals).
  • “Patterned” or “temporal” as used in the context of drug delivery means delivery of drug in a pattern, generally a substantially regular pattern, over a pre-selected period of time (e.g. , other than a period associated with, for example a bolus injection).
  • “Patterned” or “temporal” drug delivery is meant to encompass delivery of drug at an increasing, decreasing, substantially constant, or pulsatile, rate or range of rates (e.g., amount of drug per unit time, or volume of drug formulation for a unit time), and further encompasses delivery that is continuous or substantially continuous, or chronic.
  • controlled drug delivery device is meant to encompass any device wherein the release (e.g., rate, timing of release) of a drug or other desired substance contained therein is controlled by or determined by the device itself and not substantially influenced by the environment of use, or releasing at a rate that is reproducible within the environment of use.
  • substantially continuous as used in, for example, the context of “substantially continuous infusion” or “substantially continuous delivery” is meant to refer to delivery of drug in a manner that is substantially uninterrupted for a pre-selected period of drug delivery, where the quantity of drug received by the patient during any 8 hour interval in the pre-selected period never falls to zero.
  • substantially continuous drug delivery can also encompass delivery of drug at a substantially constant, pre-selected rate or range of rates (e.g., amount of drug per unit time, or volume of drug formulation for a unit time) that is substantially uninterrupted for a pre-selected period of drug delivery.
  • pirfenidone means 5-methyl-l-phenyl-2-(lH)-pyridone.
  • pirfenidone analog means any compound of Formula I, IIA or IIB below.
  • specific pirfenidone analog refers to, and is limited to, each and every pirfenidone analog shown in Table 1.
  • Type I interferon receptor agonist refers to any naturally occurring or non-naturally occurring ligand of human Type I interferon receptor, which binds to and causes signal transduction via the receptor.
  • Type I interferon receptor agonists include interferons, including naturally-occurring interferons, modified mterferons, synthetic interferons, pegylated interferons, fusion proteins comprising an interferon and a heterologous protein, shuffled interferons; antibody specific for an interferon receptor; non-peptide chemical agonists; and the like.
  • Type II interferon receptor agonist refers to any naturally- occurring or non-naturally-occurring ligand of a human Type II interferon receptor which binds to and causes signal transduction via the receptor.
  • Type II interferon receptor agonists include interferons, including naturally-occurring interferons, modified interferons, synthetic interferons, pegylated interferons, fusion proteins comprising an interferon and a heterologous protein, shuffled interferons; antibody specific for an interferon receptor; non-peptide chemical agonists; and the like.
  • the effective amounts of a Type II interferon receptor agonist and pirfenidone or a pirfenidone analog are synergistic amounts.
  • a "synergistic combination" or a "synergistic amount" of a Type II interferon re'ceptor agonist and pirfenidone or a pirfenidone analog is a combination or amount that is more effective in the therapeutic or prophylactic treatment of ALD and/or alcoholic hepatitis than the incremental improvement in treatment outcome that could be predicted or expected from a merely additive combination of (i) the therapeutic or prophylactic benefit of the Type II interferon receptor agonist when administered at that same dosage as a monotherapy and (ii) the therapeutic or prophylactic benefit of the pirfenidone or a pirfenidone analog when administered at the same dosage as a monotherapy.
  • the present invention provides a method of treating alcoholic hepatitis, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in monotherapy or in combination therapy.
  • the invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in combination therapy with a second therapeutic agent other than IFN- ⁇ .
  • the invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog, a Type II interferon receptor agonist, and a side effect management agent (e.g., a palliative agent, or an agent for the treatment, reduction, or avoidance of a side effect caused by any therapeutic agent).
  • a side effect management agent e.g., a palliative agent, or an agent for the treatment, reduction, or avoidance of a side effect caused by any therapeutic agent.
  • the invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of a TNF- ⁇ antagonist, which therapy further includes administration of an effective amount of a Type II interferon receptor agonist, and/or pirfenidone or a pirfenidone analog.
  • alcoholic liver disease includes hepatic steatosis, alcoholic hepatitis, hepatic fibrosis, and hepatic cirrhosis, which occur as the result of chronic alcohol abuse.
  • ALD alcoholic liver disease
  • the present invention provides methods of treating ALD, involving administering one or more therapeutic agents, where the monotherapy or combination therapy is effective to ameliorate one or more of hepatic steatosis, alcoholic hepatitis, hepatic fibrosis, and hepatic cirrhosis.
  • the present invention provides method of treating alcoholic hepatitis, involving administering one or more therapeutic agents, where the monotherapy or combination therapy is effective to ameliorate alcoholic hepatitis.
  • Non-Fibrotic Alcoholic Hepatitis [0030]
  • the present invention provides a method of treating early stage ALD, e.g., treating alcoholic hepatitis in the absence of hepatic fibrosis ("non-fibrotic alcoholic hepatitis”), the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in monotherapy or in combination therapy.
  • an "effective amount" of pirfenidone or a pirfenidone analog is an amount or a dosage that, when administered to an individual in monotherapy, is effective to treat non-fibrotic alcoholic hepatitis, e.g., is effective to reduce at least one symptom and/or histological characteristic of alcoholic hepatitis.
  • an effective amount of pirfenidone or a pirfenidone analog is an amount that is effective to cause a reduction in one or more symptoms of non-fibrotic alcoholic hepatitis of at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90% or more, compared with the level or degree of the symptom in the absence of treatment with pirfenidone or pirfenidone analog.
  • Symptoms of non-fibrotic alcoholic hepatitis include fever, jaundice, and tender hepatomegaly.
  • an effective amount of pirfenidone or a pirfenidone analog is an amount that is effective to cause a reduction in one or more histological characteristics of non- fibrotic alcoholic hepatitis of at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90% or more, compared with the histological characteristic in the absence of treatment with pirfenidone or a pirfenidone analog.
  • Histological characteristics of non-fibrotic alcoholic hepatitis include hepatocellular necrosis, alcoholic Mallory's hyaline bodies, and inflammatory lesions with infiltrating polymorphonuclear leukocytes and lymphocytes.
  • an effective amount of pirfenidone or a pirfenidone analog is an amount that is effective to increase an index of liver function by at least about 10%, at least about 20%, at least about 25%, at least about 30%), at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%, or more, compared to the index of liver function in an untreated individual, or in a placebo-treated individual.
  • Those skilled in the art can readily measure such indices of liver function, using standard assay methods, many of which are commercially available, and are used routinely in clinical settings.
  • a subject method for treating non-fibrotic alcoholic hepatitis comprises administering an effective amount of pirfenidone or a pirfenidone analog, and an effective amount of at least a second therapeutic agent.
  • Suitable second therapeutic agents include, but are not limited to a Type II interferon receptor agonist, and a TNF antagonist.
  • "At least a second therapeutic agent" includes: a Type II interferon receptor agonist; a TNF antagonist; a Type I interferon receptor agonist and a Type II interferon receptor agonist; a Type II interferon receptor agonist and a TNF antagonist; and a Type II interferon receptor agonist, a Type I interferon receptor agonist and a TNF antagonist.
  • "effective amounts" of pirfenidone or a pirfenidone analog and at least a second therapeutic agent are combined amounts that, when administered to an individual in combination therapy, are effective to treat non-fibrotic alcoholic hepatitis, e.g., is effective to reduce at least one symptom and/or histological characteristic of alcoholic hepatitis.
  • "effective amounts" of pirfenidone or a pirfenidone analog and at least a second therapeutic agent are combined amounts that, when administered to an individual in combination therapy, are effective to cause a reduction in one or more symptoms of non-fibrotic alcoholic hepatitis of at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%), or at least about 90% or more, compared with the level or degree of the symptom in the absence of treatment with the combination therapy.
  • Symptoms of non-fibrotic alcoholic hepatitis include fever, jaundice, and tender hepatomegaly.
  • "effective amounts" of pirfenidone or a pirfenidone analog and at least a second therapeutic agent are combined amounts that, when administered to an individual in combination therapy, are effective to cause a reduction in one or more histological characteristics of non-fibrotic alcoholic hepatitis of at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%>, at least about 70%, at least about 80%, or at least about 90% or more, compared with the histological characteristic in the absence of treatment with the combination therapy.
  • Histological characteristics of non-fibrotic alcoholic hepatitis include hepatocellular necrosis, alcoholic Mallory's hyaline bodies, and inflammatory lesions with infiltrating polymorphonuclear leukocytes and lymphocytes.
  • "effective amounts" of pirfenidone or a pirfenidone analog and at least a second therapeutic agent are combined amounts that, when administered to an individual in combination therapy, are effective to increase an index of liver function by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 10%, at least about 75%, or at least about 80%, or more, compared to the index of liver function in an untreated individual, or in a placebo-treated individual.
  • Those skilled in the art can readily measure such indices of liver function, using standard assay methods, many of which are commercially available, and are used routinely in clinical settings. Alcoholic liver disease
  • the present invention provides methods of treating alcoholic liver disease, including reducing alcoholic hepatitis, reducing clinical liver fibrosis, reducing the likelihood that liver fibrosis will occur, reducing a parameter associated with liver fibrosis, and reducing liver cirrhosis.
  • the methods involve administering an effective amount of pirfenidone or a pirfenidone analog in monotherapy. In some embodiments, the methods involve administering an effective amount of pirfenidone or a pirfenidone analog in combination therapy with an effective amount of a second therapeutic agent other than IFN- ⁇ (e.g., where the second therapeutic agent is a TNF antagonist).
  • a second therapeutic agent other than IFN- ⁇ (e.g., where the second therapeutic agent is a TNF antagonist).
  • the methods involve administering an effective amount of pirfenidone or a pirfenidone analog, an effective amount of a Type II interferon receptor agonist, and a side effect management agent that reduces at least one side effect of a pirfenidone or a pirfenidone analog and/or a Type II interferon receptor agonist.
  • the methods involve co-administering an effective amount a TNF antagonist and an effective amount of a Type II interferon receptor agonist; in some of these embodiments, the methods further involve administering pirfenidone or a pirfenidone analog.
  • the methods involve administering an effective amount of a TNF antagonist, an effective amount of a Type II interferon receptor agonist, and an effective amount of a Type I interferon receptor agonist; in some of these embodiments, the methods further involve administering pirfenidone or a pirfenidone analog. In some embodiments, the methods involve administering an effective amount of a TNF antagonist and an effective amount of pirfenidone or a pirfenidone analog. Of particular interest in many embodiments is treatment of humans.
  • Alcoholic liver fibrosis is a precursor to liver cirrhosis. Accordingly, the present invention further provides methods of reducing the likelihood that an individual will develop liver cirrhosis. Alcoholic liver fibrosis is a precursor to the complications associated with liver cirrhosis, such as portal hypertension, progressive liver insufficiency, and hepatocellular carcinoma. A reduction in liver fibrosis thus reduces the incidence of such complications. Accordingly, the present invention further provides methods of reducing the likelihood that an individual will develop complications associated with cirrhosis of the liver.
  • a therapeutic agent e.g., a Type II interferon receptor agonist, a Type I interferon receptor agonist, a TNF- ⁇ antagonist, pirfenidone or a pirfenidone analog, or any of the above-mentioned combinations thereof
  • a therapeutic agent e.g., a Type II interferon receptor agonist, a Type I interferon receptor agonist, a TNF- ⁇ antagonist, pirfenidone or a pirfenidone analog, or any of the above-mentioned combinations thereof
  • a therapeutic agent e.g., a Type II interferon receptor agonist, a Type I interferon receptor agonist, a TNF- ⁇ antagonist, pirfenidone or a pirfenidone analog, or any of the above-mentioned combinations thereof
  • a therapeutic agent e.g., a Type II interferon receptor agonist, a Type I interferon receptor agonist, a T
  • the invention also provides a method for treatment of alcoholic liver fibrosis in an individual comprising one or more therapeutic agents as described above in monotherapy or in combination therapy in amounts that are effective for prophylaxis or therapy of liver fibrosis in the individual, e.g., increasing the probability of survival, reducing the risk of death, ameliorating the disease burden or slowing the progression of disease in the individual.
  • Whether a subject monotherapy or combination therapy is effective in reducing alcoholic liver fibrosis can be determined by any of a number of well-established techniques for measuring liver fibrosis and liver function. Whether liver fibrosis is reduced is determined by analyzing a liver biopsy sample.
  • An analysis of a liver biopsy comprises assessments of two major components: necroinflammation assessed by "grade” as a measure of the severity and ongoing disease activity, and the lesions of fibrosis and parenchymal or vascular remodeling as assessed by "stage” as being reflective of long-term disease progression. See, e.g., Brunt (2000) Hepatol. 31:241-246; and METAVIR (1994) Hepatology 20:15-20. Based on analysis of the liver biopsy, a score is assigned. A number of standardized scoring systems exist which provide a quantitative assessment of the degree and severity of fibrosis. These include the METAVIR, Knodell, Scheuer, Ludwig, and Ishak scoring systems.
  • the METAVIR scoring system is based on an analysis of various features of a liver biopsy, including fibrosis (portal fibrosis, centrilobular fibrosis, and cirrhosis); necrosis (piecemeal and lobular necrosis, acidophilic retraction, and ballooning degeneration); inflammation (portal tract inflammation, portal lymphoid aggregates, and distribution of portal inflammation); bile duct changes; and the Knodell index (scores of periportal necrosis, lobular necrosis, portal inflammation, fibrosis, and overall disease activity).
  • each stage in the METAVIR system is as follows: score: 0, no fibrosis; score: 1, stellate enlargement of portal tract but without septa formation; score: 2, enlargement of portal tract with rare septa formation; score: 3, numerous septa without cirrhosis; and score: 4, cirrhosis.
  • Knodell's scoring system also called the Hepatitis Activity Index, classifies specimens based on scores in four categories of histologic features: I. Periportal and/or bridging necrosis; II. Intralobular degeneration and focal necrosis; III. Portal inflammation ; and IV. Fibrosis.
  • scores are as follows: score: 0, no fibrosis; score: 1, mild fibrosis (fibrous portal expansion); score: 2, moderate fibrosis; score: 3, severe fibrosis (bridging fibrosis); and score: 4, cirrhosis. The higher the score, the more severe the liver tissue damage.
  • the Ishak scoring system is described in Ishak (1995) J Hepatol. 22:696-699. Stage 0, No fibrosis; Stage 1, Fibrous expansion of some portal areas, with or without short fibrous septa; stage 2, Fibrous expansion of most portal areas, with or without short fibrous septa; stage 3, Fibrous expansion of most portal areas with occasional portal to portal (P-P) bridging; stage 4, Fibrous expansion of portal areas with marked bridging (P-P) as well as portal-central (P-C); stage 5, Marked bridging (P-P and/or P-C) with occasional nodules (incomplete cirrhosis); stage 6, Cirrhosis, probable or definite .
  • the benefit of anti-fibrotic therapy can also be measured and assessed by using the Child-Pugh scoring system which comprises a multicomponent point system based upon abnormalities in serum bilirubin level, serum albumin level, prothrombin time, the presence and severity of ascites, and the presence and severity of encephalopathy. Based upon the presence and severity of abnormality of these parameters, patients may be placed in one of three categories of increasing severity of clinical disease: A, B, or C.
  • therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that effects a change of one unit or more in the fibrosis stage based on pre- and post- therapy liver biopsies.
  • therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) reduce liver fibrosis by at least one unit in the METAVIR, the Knodell, the Scheuer, the Ludwig, or the Ishak scoring system.
  • Secondary, or indirect, indices of liver function can also be used to evaluate the efficacy of treatment with the subject therapy. Morphometric computerized semi-automated assessment of the quantitative degree of liver fibrosis based upon specific staining of collagen and/or serum markers of liver fibrosis can also be measured as an indication of the efficacy of a subject treatment method. Secondary indices of liver function include, but are not limited to, serum transaminase levels, prothrombin time, bilirubin, platelet count, portal pressure, albumin level, and assessment of the Child-Pugh score.
  • therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that are effective to increase an index of liver function by at least about 10%, at least about 20%), at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%>, at least about 65%, at least about 70%, at least about 75%, or at least about 80%>, or more, compared to the index of liver function in an untreated individual, or in a placebo-treated individual.
  • Those skilled in the art can readily measure such indices of liver function, using standard assay methods, many of which are commercially available, and are used routinely in clinical settings.
  • Serum markers of liver fibrosis can also be measured as an indication of the efficacy of a subject treatment method.
  • Serum markers of liver fibrosis include, but are not limited to, hyaluronate, N-terminal procollagen III peptide, 7S domain of type IV collagen, C-terminal procollagen I peptide, and laminin.
  • Additional biochemical markers of liver fibrosis include ⁇ - 2-macroglobulin, haptoglobin, gamma globulin, apolipoprotein A, and gamma glutamyl transpeptidase.
  • therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that are effective to reduce a serum level of a marker of liver fibrosis by at least about 10%), at least about 20%, at least about 25%>, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%>, at least about 55%>, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%, or more, compared to the level of the marker in an untreated individual, or in a placebo-treated individual.
  • a dosage in a subject monotherapy or a combined dosage in a subject combination therapy that are effective to reduce a serum level of a marker of liver fibrosis by at least about 10%
  • ELISA enzyme-linked immunosorbent assays
  • radioimmunoassays radioimmunoassays
  • Quantitative tests of functional liver reserve can also be used to assess the efficacy of treatment with the subject therapy. These include: indocyanine green clearance (ICG), galactose elimination capacity (GEC), aminopyrine breath test (ABT), antipyrine clearance, monoethylglycine-xylidide (MEG-X) clearance, and caffeine clearance.
  • a "complication associated with cirrhosis of the liver” refers to a disorder that is a sequellae of decompensated liver disease, i.e., or occurs subsequently to and as a result of development of liver fibrosis, and includes, but is not limited to, development of ascites, variceal bleeding, portal hypertension, jaundice, progressive liver insufficiency, encephalopathy, hepatocellular carcinoma, liver failure requiring liver transplantation, and liver-related mortality.
  • therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that is effective in reducing the incidence of (e.g., the likelihood that an individual will develop) a disorder associated with cirrhosis of the liver by at least about 10%, at least about 20%, at least about 25%, at least about 30%>, at least about 35%, at least about 40%, at least about 45%>, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%, or more, compared to an untreated individual, or in a placebo-treated individual.
  • a dosage e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy
  • Whether a subject monotherapy or combination therapy is effective in reducing the incidence of a disorder associated with cirrhosis of the liver can readily be determined by those skilled in the art.
  • therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that is effective in increasing liver function.
  • Liver functions include, but are not limited to, synthesis of proteins such as serum proteins (e.g., albumin, clotting factors, alkaline phosphatase, aminotransferases (e.g., alanine transaminase, aspartate transaminase), 5'-nucleosidase, ⁇ - glutaminyltranspeptidase, etc.), synthesis of bilirubin, synthesis of cholesterol, and synthesis of bile acids; a liver metabolic function, including, but not limited to, carbohydrate metabolism, amino acid and ammonia metabolism, hormone metabolism, and lipid metabolism; detoxification of exogenous drugs; a hemodynamic function, including splanchnic and portal hemodynamics; and the like.
  • proteins such as serum proteins (e.g., albumin, clotting factors, alkaline phosphatase, aminotransferases (e.g., alanine transaminase, aspartate transaminase), 5'
  • liver function is increased is readily ascertainable by those skilled in the art, using well-established tests of liver function.
  • markers of liver function such as albumin, alkaline phosphatase, alanine transaminase, aspartate transaminase, bilirubin, and the like, can be assessed by measuring the level of these markers in the serum, using standard immunological and enzymatic assays.
  • Splanchnic circulation and portal hemodynamics can be measured by portal wedge pressure and/or resistance using standard methods.
  • Metabolic functions can be measured by measuring the level of ammonia in the serum.
  • Whether serum proteins normally secreted by the liver are in the normal range can be determined by measuring the levels of such proteins, using standard immunological and enzymatic assays. Those skilled in the art know the normal ranges for such serum proteins. The following are non-limiting examples.
  • the normal range of alanine transaminase is from about 7 to about 56 units per liter of serum.
  • the normal range of aspartate transaminase is from about 5 to about 40 units per liter of serum.
  • Bilirubin is measured using standard assays. Normal bilirubin levels are usually less than about 1.2 mg/dL.
  • Serum albumin levels are measured using standard assays. Normal levels of serum albumin are in the range of from about 35 to about 55 g/L.
  • Prolongation of prothrombin time is measured using standard assays. Normal prothrombin time is less than about 4 seconds longer than control.
  • therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that is effective to increase liver function by at least about 10%), at least about 20%), at least about 30%, at least about 40%, at least about 50%>, at least about 60%, at least about 70%, at least about 80%, or more, compared to an untreated individual, or in a placebo-treated individual.
  • a dosage in a subject monotherapy or a combined dosage in a subject combination therapy that is effective to increase liver function by at least about 10%), at least about 20%), at least about 30%, at least about 40%, at least about 50%>, at least about 60%, at least about 70%, at least about 80%, or more, compared to an untreated individual, or in a placebo-treated individual.
  • therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that is effective to reduce an elevated level of a serum marker of liver function by at least about 10%>, at least about 20%, at least about 30%, at least about 40%, at least about 50%), at least about 60%>, at least about 70%, at least about 80%, or more, or to reduce the level of the serum marker of liver function to within a normal range.
  • therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) effective to increase a reduced level of a serum marker of liver function by at least about 10%, at least about 20%), at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%), at least about 80%, or more, or to increase the level of the serum marker of liver function to within a normal range.
  • dosages e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy
  • Type I interferon receptor agonists include an IFN- ⁇ ; an IFN- ⁇ ; an IFN-tau; an IFN- ⁇ ; antibody agonists specific for a Type I interferon receptor; and any other agonist of Type I interferon receptor, including non-polypeptide agonists.
  • Interferon-Alpha Interferon-Alpha
  • IFN- ⁇ any known IFN- ⁇ can be used in the instant invention.
  • interferon-alpha refers to a family of related polypeptides that inhibit viral replication and cellular proliferation and modulate immune response.
  • IFN- ⁇ includes naturally occurring IFN- ⁇ ; synthetic IFN- ⁇ ; derivatized IF ⁇ - ⁇ (e.g., PEGylated IFN- ⁇ , glycosylated IFN- ⁇ , and the like); and analogs of naturally occurring or synthetic IFN- ⁇ ; essentially any IFN- ⁇ that has antiviral properties, as described for naturally occurring IFN- ⁇ .
  • Suitable alpha interferons include, but are not limited to, naturally-occurring IFN- ⁇ (including, but not limited to, naturally occurring IFN- ⁇ 2a, IFN- ⁇ 2b); recombinant interferon alpha-2b such as Intron-A interferon available from Schering Corporation, Kenilworth, N.J.; recombinant interferon alpha-2a such as Roferon interferon available from Hoffmann-La Roche, Nutley, N.
  • interferon alpha-2C such as Berofor alpha 2 interferon available from Boehringer Ingelheim Pharmaceutical, Inc., Ridgefield, Conn.
  • interferon alpha- nl a purified blend of natural alpha interferons such as Sumiferon available from Sumitomo, Japan or as Wellferon interferon alpha-nl (INS) available from the Glaxo-Wellcome Ltd., London, Great Britain
  • interferon alpha-n3 a mixture of natural alpha interferons made by Interferon Sciences and available from the Purdue Frederick Co., Norwalk, Conn., under the Alferon Tradename.
  • IFN- ⁇ also encompasses consensus IFN- ⁇ .
  • Consensus IF ⁇ - ⁇ (also referred to as “CIFN” and “IFN-con” and “consensus interferon”) encompasses but is not limited to the amino acid sequences designated IFN-coni, IFN-con 2 and IFN-con 3 which are disclosed in U.S. Pat. Nos. 4,695,623 and 4,897,471; and consensus interferon as defined by determination of a consensus sequence of naturally occurring interferon alphas (e.g., Infergen®, InterMune, Inc., Brisbane, Calif).
  • IFN-coni is the consensus interferon agent in the Infergen® alfacon-1 product.
  • IFN-con The Infergen® consensus interferon product is referred to herein by its brand name (Infergen®) or by its generic name (interferon alfacon-1). DNA sequences encoding IFN-con may be synthesized as described in the aforementioned patents or other standard methods. Use of CIFN is of particular interest.
  • IFN- ⁇ fusion polypeptides comprising an IFN- ⁇ and a heterologous polypeptide.
  • IFN- ⁇ fusion polypeptides include, but are not limited to, Albuferon-alphaTM (a fiision product of human albumin and IFN- ⁇ ; Human Genome Sciences; see, e.g., Osborn et al. (2002) J Pharmacol. Exp. Therap. 303:540-548).
  • gene-shuffled forms of IFN- ⁇ See., e.g., Masci et al. (2003) Curr. Oncol. Rep. 5:108-113. PEGylated Interferon-Alpha
  • IFN- ⁇ also encompasses derivatives of IFN- ⁇ that are derivatized (e.g., are chemically modified) to alter certain properties such as serum half-life.
  • IFN- ⁇ includes glycosylated IFN- ⁇ ; IF ⁇ - ⁇ derivatized with polyethylene glycol ("PEGylated IFN- ⁇ "); and the like. PEGylated IFN- ⁇ , and methods for making same, is discussed in, e.g., U.S. Patent Nos. 5,382,657; 5,981,709; and 5,951,974.
  • PEGylated IFN- ⁇ encompasses conjugates of PEG and any of the above-described IFN- ⁇ molecules, including, but not limited to, PEG conjugated to interferon alpha-2a (Roferon, Hoffman La-Roche, Nutley, N. J.), interferon alpha 2b (Intron, Schering-Plough, Madison, N. J.), interferon alpha-2c (Berofor Alpha, Boehringer Ingelheim, Ingelheim, Germany); and consensus interferon as defined by determination of a consensus sequence of naturally occurring interferon alphas (Infergen®, InterMune, Inc., Brisbane, Calif).
  • any of the above-mentioned IFN- ⁇ polypeptides can be modified with one or more polyethylene glycol moieties, i.e., PEGylated.
  • the PEG molecule of a PEGylated IFN- ⁇ polypeptide is conjugated to one or more amino acid side chains of the IFN- ⁇ polypeptide.
  • the PEGylated IFN- ⁇ contains a PEG moiety on only one amino acid.
  • the PEGylated IFN- ⁇ contains a PEG moiety on two or more amino acids, e.g., the IFN- ⁇ contains a PEG moiety attached to two, three, four, five, six, seven, eight, nine, or ten different amino acid residues.
  • IFN- ⁇ may be coupled directly to PEG (i.e., without a linking group) through an amino group, a sulfhydryl group, a hydroxyl group, or a carboxyl group.
  • the PEGylated IFN- ⁇ is PEGylated at or near the amino terminus ( ⁇ -terminus) of the IFN- ⁇ polypeptide, e.g., the PEG moiety is conjugated to the IFN- ⁇ polypeptide at one or more amino acid residues from amino acid 1 through amino acid 4, or from amino acid 5 through about 10.
  • the PEGylated IFN- ⁇ is PEGylated at one or more amino acid residues from about 10 to about 28.
  • the PEGylated IFN- ⁇ is PEGylated at or near the carboxyl terminus (C-terminus) of the IFN- ⁇ polypeptide, e.g., at one or more residues from amino acids 156-166, or from amino acids 150 to 155.
  • the PEGylated IFN- ⁇ is PEGylated at one or more amino acid residues at one or more residues from amino acids 100-114.
  • amino acids at which PEGylation is to be avoided include amino acid residues from amino acid 30 to amino acid 40; and amino acid residues from amino acid 113 to amino acid 149.
  • PEG is attached to IFN- ⁇ via a linking group.
  • the linking group is any biocompatible linking group, where "biocompatible" indicates that the compound or group is non-toxic and may be utilized in vitro or in vivo without causing injury, sickness, disease, or death.
  • PEG can be bonded to the linking group, for example, via an ether bond, an ester bond, a thiol bond or an amide bond.
  • Suitable biocompatible linking groups include, but are not limited to, an ester group, an amide group, an imide group, a carbamate group, a carboxyl group, a hydroxyl group, a carbohydrate, a succinimide group (including, for example, succinimidyl succinate (SS), succinimidyl propionate (SPA), succinimidyl butanoate (SBA), succinimidyl carboxymethylate (SCM), succinimidyl succinamide (SSA) or N-hydroxy succinimide (NHS)), an epoxide group, an oxycarbonylimidazole group (including, for example, carbonyldimidazole (GDI)), a nitro phenyl group (including, for example, nitrophenyl carbonate ( ⁇ PC) or trichlorophenyl carbonate (TPC)), a trysylate group, an aldehyde group, an isocyanate group, a vinylsulfone group, a
  • succinimidyl propionate (SPA) and succinimidyl butanoate (SBA) ester-activated PEGs are described in U.S. Pat. No. 5,672,662 (Harris, et al.) and WO 97/03106.
  • Pegylated IFN- ⁇ and methods for making same, is discussed in, e.g., U.S. Patent Nos. 5,382,657; 5,981,709; 5,985,265; and 5,951,974.
  • Pegylated IFN- ⁇ encompasses conjugates of PEG and any of the above-described IFN- ⁇ molecules, including, but not limited to, PEG conjugated to interferon alpha-2a (Roferon, Hoffman LaRoche, Nutley, N. J.), where PEGylated Roferon is known as Pegasys (Hoffman LaRoche); interferon alpha 2b (Intron, Schering-Plough, Madison, N.
  • PEGylated Intron is known as PEG-Intron (Schering- Plough); interferon alpha-2c (Berofor Alpha, Boeliringer Ingelheim, Ingelheim, Germany); and consensus interferon (CIFN) as defined by determination of a consensus sequence of naturally occurring interferon alphas (Infergen®, InterMune, Inc., Brisbane, Calif), where PEGylated Infergen is referred to as PEG-Infergen.
  • the PEG is a monomethoxyPEG molecule that reacts with primary amine groups on the IFN- ⁇ polypeptide.
  • Methods of modifying polypeptides with monomethoxy PEG via reductive alkylation are known in the art. See, e.g., Chamow et al. (1994) Bioconj. Chem. 5:133-140.
  • PEG is linked to IFN- ⁇ via an SPA linking group.
  • SPA esters of PEG, and methods for making same, are described in U.S. Patent No. 5,672,662.
  • SPA linkages provide for linkage to free amine groups on the IFN- ⁇ polypeptide.
  • a PEG molecule is covalently attached via a linkage that comprises an amide bond between a propionyl group of the PEG moiety and the epsilon amino group of a surface-exposed lysine residue in the IFN- ⁇ polypeptide.
  • a linkage that comprises an amide bond between a propionyl group of the PEG moiety and the epsilon amino group of a surface-exposed lysine residue in the IFN- ⁇ polypeptide.
  • Such a bond can be formed, e.g., by condensation of an ⁇ -methoxy, omega propanoic acid activated ester of PEG (mPEGspa).
  • one monopegylated CIFN conjugate preferred for use herein has a linear PEG moiety of about 30 kD attached via a covalent linkage to the CIFN polypeptide, where the covalent linkage is an amide bond between a propionyl group of the PEG moiety and the epsilon amino group of a surface-exposed lysine residue in the CIFN polypeptide, where the surface-exposed lysine residue is chosen from lys , lys , lys , lys , lys , and the amide bond is formed by condensation of an ⁇ - methoxy, omega propanoic acid activated ester of PEG.
  • Polyethylene glycol Polyethylene glycol
  • Polyethylene glycol suitable for conjugation to an IFN- ⁇ polypeptide is soluble in water at room temperature, and has the general formula R(O-CH 2 -CH 2 ) n O-R, where R is hydrogen or a protective group such as an alkyl or an alkanol group, and where n is an integer from 1 to 1000. Where R is a protective group, it generally has from 1 to 8 carbons.
  • PEG has at least one hydroxyl group, e.g., a terminal hydroxyl group, which hydroxyl group is modified to generate a functional group that is reactive with an amino group, e.g., an epsilon amino group of a lysine residue, a free amino group at the N- terminus of a polypeptide, or any other amino group such as an amino group of asparagine, glutamine, arginine, or histidine.
  • an amino group e.g., an epsilon amino group of a lysine residue, a free amino group at the N- terminus of a polypeptide, or any other amino group such as an amino group of asparagine, glutamine, arginine, or histidine.
  • PEG is derivatized so that it is reactive with free carboxyl groups in the IFN- ⁇ polypeptide, e.g., the free carboxyl group at the carboxyl terminus of the IFN- ⁇ polypeptide.
  • Suitable derivatives of PEG that are reactive with the free carboxyl group at the carboxyl-terminus of IFN- ⁇ include, but are not limited to PEG-amine, and hydrazine derivatives of PEG (e.g., PEG-NH-NH 2 ).
  • PEG is derivatized such that it comprises a terminal thiocarboxylic acid group, -COSH, which selectively reacts with amino groups to generate amide derivatives.
  • -SH a terminal thiocarboxylic acid group
  • selectivity of certain amino groups over others is achieved.
  • -SH exhibits sufficient leaving group ability in reaction with N-terminal amino group at appropriate pH conditions such that the ⁇ -amino groups in lysine residues are protonated and remain non-nucleophilic.
  • reactions under suitable pH conditions may make some of the accessible lysine residues to react with selectivity.
  • the PEG comprises a reactive ester such as an N-hydroxy succinimidate at the end of the PEG chain.
  • a reactive ester such as an N-hydroxy succinimidate at the end of the PEG chain.
  • Such an N-hydroxysuccinimidate-containing PEG molecule reacts with select amino groups at particular pH conditions such as neutral 6.5-7.5.
  • the N-terminal amino groups may be selectively modified under neutral pH conditions.
  • accessible-NH 2 groups of lysine may also react.
  • the PEG can be conjugated directly to the IFN- ⁇ polypeptide, or through a linker.
  • a linker is added to the IFN- ⁇ polypeptide, forming a linker-modified IFN- ⁇ polypeptide.
  • Such linkers provide various functionalities, e.g., reactive groups such sulfhydryl, amino, or carboxyl groups to couple a PEG reagent to the linker-modified IFN- ⁇ polypeptide.
  • the PEG conjugated to the IFN- ⁇ polypeptide is linear. In other embodiments, the PEG conjugated to the IFN- ⁇ polypeptide is branched. Branched PEG derivatives such as those described in U.S. Pat. No. 5,643,575, "star-PEG's” and multi-armed PEG's such as those described in Shearwater Polymers, Inc. catalog "Polyethylene Glycol Derivatives 1997-1998.” Star PEGs are described in the art including, e.g., in U.S. Patent No. 6,046,305.
  • PEG having a molecular weight in a range of from about 2 kDa to about 100 kDa is generally used, where the term "about,” in the context of PEG, indicates that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight.
  • PEG suitable for conjugation to IFN- ⁇ has a molecular weight of from about 2 kDa to about 5 kDa, from about 5 kDa to about 10 kDa, from about 10 kDa to about 15 IcDa, from about 15 kDa to about 20 kDa, from about 20 kDa to about 25 kDa, from about 25 IcDa to about 30 kDa, from about 30 kDa to about 40 IcDa, from about 40 kDa to about 50 IcDa, from about 50 kDa to about 60 kDa, from about 60 kDa to about 70 kDa, from about 70 IcDa to about 80 kDa, from about 80 kDa to about 90 kDa, or from about 90 kDa to about 100 kDa.
  • the PEG moiety can be attached, directly or via a linker, to an amino acid residue at or near the N-terminus, internally, or at or near the C-terminus of the IFN- ⁇ polypeptide. Conjugation can be carried out in solution or in the solid phase. N-terminal linkage
  • known methods for selectively obtaining an N-terminally chemically modified IFN- ⁇ are used.
  • a method of protein modification by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminus) available for derivatization in a particular protein can be used.
  • substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved.
  • the reaction is performed at pH which allows one to take advantage of the pK a differences between the ⁇ - amino groups of the lysine residues and that of the ⁇ -amino group of the N-terminal residue of the protein.
  • N-terminal-specific coupling procedures such as described in U.S. Patent No. 5,985,265 provide predominantly monoPEGylated products.
  • the purification procedures aimed at removing the excess reagents and minor multiply PEGylated products remove the N-terminal blocked polypeptides.
  • such processes lead to significant increases in manufacturing costs.
  • examination of the structure of the well-characterized Infergen® Alfacon-1 CIFN polypeptide amino acid sequence reveals that the clipping is approximate 5% at the carboxyl terminus and thus there is only one major C- terminal sequence.
  • N-terminally PEGylated IFN- ⁇ is not used; instead, the IFN- ⁇ polypeptide is C-terminally PEGylated.
  • a PEG reagent that is selective for the C-terminal can be prepared wit or without spacers.
  • polyethylene glycol modified as methyl ether at one end and having an amino function at the other end may be used as the starting material.
  • Preparing or obtaining a water-soluble carbodiimide as the condensing agent can be carried out.
  • Coupling IFN- ⁇ e.g., Infergen® Alfacon-1 CIFN or consensus interferon
  • a water-soluble carbodiimide as the condensing reagent is generally carried out in aqueous medium with a suitable buffer system at an optimal pH to effect the amide linkage.
  • a high molecular weight PEG can be added to the protein covalently to increase the molecular weight.
  • ED AC ED AC
  • l-ethyl-3- (3-dimethylaminopropyl) carbodiimide a suitable reagent.
  • the water solubility of ED AC allows for direct addition to a reaction without the need for prior organic solvent dissolution.
  • Excess reagent and the isourea formed as the by-product of the cross-linking reaction are both water-soluble and may easily be removed by dialysis or gel filtration.
  • a concentrated solution of ED AC in water is prepared to facilitate the addition of a small molar amount to the reaction. The stock solution is prepared and used immediately in view of the water labile nature of the reagent.
  • the optimal reaction medium to be in pH range between 4.7 and 6.0. However the condensation reactions do proceed without significant losses in yields up to pH 7.5. Water may be used as solvent.
  • the medium will be 2-(N-morpholino)efhane sulfonic acid buffer pre-titrated to pH between 4.7 and 6.0.
  • 0.1M phosphate in the pH 7-7.5 may also be used in view of the fact that the product is in the same buffer.
  • the ratios of PEG amine to the IF ⁇ - ⁇ molecule is optimized such that the C- terminal carboxyl residue(s) are selectively PEGylated to yield monoPEGylated derivative(s).
  • PEG amine has been mentioned above by name or structure, such derivatives are meant to be exemplary only, and other groups such as hydrazine derivatives as in PEG-NH-NH 2 which will also condense with the carboxyl group of the IFN- ⁇ protein, can also be used.
  • the reactions can also be conducted on solid phase.
  • Polyethylene glycol can be selected from list of compounds of molecular weight ranging from 300-40000. The choice of the various polyethylene glycols will also be dictated by the coupling efficiency and the biological performance of the purified derivative in vitro and in vivo i.e., circulation times, anti viral activities etc.
  • suitable spacers can be added to the C-terminal of the protein.
  • the spacers may have reactive groups such as SH, NH 2 or COOH to couple with appropriate PEG reagent to provide the high molecular weight IFN- ⁇ derivatives.
  • a combined solid/solution phase methodology can be devised for the preparation of C-terminal pegylated interferons. For example, the C-terminus of IFN- ⁇ is extended on a solid phase using a Gly-Gly-Cys-NH 2 spacer and then monopegylated in solution using activated dithiopyridyl-PEG reagent of appropriate molecular weights.
  • C-terminal PEGylation Another method of achieving C-terminal PEGylation is as follows. Selectivity of C- terminal PEGylation is achieved with a sterically hindered reagent which excludes reactions at carboxyl residues either buried in the helices or internally in IFN- ⁇ .
  • a sterically hindered reagent which excludes reactions at carboxyl residues either buried in the helices or internally in IFN- ⁇ .
  • one such reagent could be a branched chain PEG ⁇ 40kd in molecular weight and this agent could be synthesized as follows:
  • OH 3 C-(CH 2 CH 2 O)n-CH 2 CH 2 NH 2 + Glutamic Acid i.e., HOCO-CH 2 CH 2 CH( ⁇ H2 COOH is condensed with a suitable agent e.g., dicyclohexyl carbodiimide or water-soluble ED AC to provide the branched chain PEG agent OH 3 C-(CH 2 CH 2 O) n - CH 2 CH 2 NHCOCH(NH 2 )CH 2 OCH 3 -(CH 2 CH 2 O) n -CH 2 CH 2 NHCOCH 2 .
  • a suitable agent e.g., dicyclohexyl carbodiimide or water-soluble ED AC
  • This reagent can be used in excess to couple the amino group with the free and flexible carboxyl group of IFN- ⁇ to form the peptide bond.
  • PEGylated IFN- ⁇ is separated from unPEGylated IFN- ⁇ using any known method, including, but not limited to, ion exchange chromatography, size exclusion chromatography, and combinations thereof.
  • the products are first separated by ion exchange cl romatography to obtain material having a charge characteristic of monoPEGylated material (other multi- PEGylated material having the same apparent charge may be present), and then the monoPEGylated materials are separated using size exclusion cliromatography.
  • interferon-beta includes IFN- ⁇ polypeptides that are naturally occurring; non-naturally-occurring IFN- ⁇ polypeptides; and analogs of naturally occurring or non-naturally occurring IFN- ⁇ that retain antiviral activity of a parent naturally-occurring or non-naturally occurring IFN- ⁇ .
  • beta interferons can be administered in conjunction with a subject method.
  • Suitable beta interferons include, but are not limited to, naturally-occurring IFN- ⁇ ; IFN- ⁇ la, e.g., Avonex® (Biogen, Inc.), and Rebif® (Serono, SA); IFN- ⁇ lb (Betaseron®; Berlex); and the like.
  • the IFN- ⁇ formulation may comprise an N-blocked species, wherein the N-terminal amino acid is acylated with an acyl group, such as a formyl group, an acetyl group, a malonyl group, and the like. Also suitable for use is a consensus IFN- ⁇ .
  • IFN- ⁇ polypeptides can be produced by any known method. DNA sequences encoding IFN- ⁇ may be synthesized using standard methods. In many embodiments, IFN- ⁇ polypeptides are the products of expression of manufactured DNA sequences transformed or transfected into bacterial hosts, e.g., E. coli, or in eukaryotic host cells (e.g., yeast; mammalian cells, such as CHO cells; and the like). In these embodiments, the IFN- ⁇ is "recombinant IFN- ⁇ .” Where the host cell is a bacterial host cell, the IFN- ⁇ is modified to comprise an N- terminal methionine.
  • IFN- ⁇ as described herein may comprise one or more modified amino acid residues, e.g., glycosylations, chemical modifications, and the like.
  • IFN-tau may comprise one or more modified amino acid residues, e.g., glycosylations, chemical modifications, and the like.
  • the tenri interferon-tau includes IFN-tau polypeptides that are naturally occurring; non- naturally-occurring IFN-tau polypeptides; and analogs of naturally occurring or non-naturally occurring IFN-tau that retain antiviral activity of a parent naturally-occurring or non-naturally occurring IFN-tau.
  • Suitable tau interferons include, but are not limited to, naturally-occurring IFN-tau; Tauferon® (Pepgen Corp.); and the like.
  • IFN-tau may comprise an amino acid sequence as set forth in any one of GenBank Accession Nos. P15696; P56828; P56832; P56829; P56831; Q29429; Q28595; Q28594; S08072; Q08071; Q08070; Q08053; P56830; P28169; P28172; and P28171.
  • the sequence of any known IFN-tau polypeptide may be altered in various ways known in the art to generate targeted changes in sequence.
  • a variant polypeptide will usually be substantially similar to the sequences provided herein, i.e. will differ by at least one amino acid, and may differ by at least two but not more than about ten amino acids.
  • sequence changes may be substitutions, insertions or deletions.
  • Conservative amino acid substitutions typically include substitutions within the following groups: (glycine, alanine); (valine, isoleucine, leucine); (aspartic acid, glutamic acid); (asparagine, glutamine); (serine, threonine); (lysine, arginine); or (phenylalanine, tyrosine).
  • Modifications of interest that may or may not alter the primary amino acid sequence include chemical derivatization of polypeptides, e.g., acetylation, or carboxylation; changes in amino acid sequence that introduce or remove a glycosylation site; changes in amino acid sequence that make the protein susceptible to PEGylation; and the like. Also included are modifications of glycosylation, e.g. those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g. by exposing the polypeptide to enzymes that affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes.
  • the IFN-tau fonnulation may comprise an N-blocked species, wherein the N-terminal amino acid is acylated with an acyl group, such as a formyl group, an acetyl group, a malonyl group, and the like. Also suitable for use is a consensus IFN-tau.
  • IFN-tau polypeptides can be produced by any known method. DNA sequences encoding IFN-tau may be synthesized using standard methods. In many embodiments, IFN-tau polypeptides are the products of expression of manufactured DNA sequences transformed or transfected into bacterial hosts, e.g., E. coli, or in eukaryotic host cells (e.g., yeast; mammalian cells, such as CHO cells; and the like). In these embodiments, the IFN-tau is "recombinant IF ⁇ -tau.” Where the host cell is a bacterial host cell, the IFN-tau is modified to comprise an N-terminal methionine.
  • IFN-tau as described herein may comprise one or more modified amino acid residues, e.g., glycosylations, chemical modifications, and the like.
  • IFN- ⁇ may comprise one or more modified amino acid residues, e.g., glycosylations, chemical modifications, and the like.
  • interferon-omega includes IFN- ⁇ polypeptides that are naturally occurring; non-naturally-occurring IFN- ⁇ polypeptides; and analogs of naturally occurring or non-naturally occurring IFN- ⁇ that retain antiviral activity of a parent naturally-occurring or non-naturally occurring IFN- ⁇ .
  • Suitable omega interferon can be administered in a subject method.
  • Suitable IFN- ⁇ include, but are not limited to, naturally-occurring IFN- ⁇ ; recombinant IFN- ⁇ , e.g., Biomed 510 (BioMedicines); and the like.
  • IFN- ⁇ may comprise an amino acid sequence as set forth in GenBank Accession No. NP_002168; or AAA70091.
  • the sequence of any known IFN- ⁇ polypeptide may be altered in various ways known in the art to generate targeted changes in sequence.
  • a variant polypeptide will usually be substantially similar to the sequences provided herein, i.e. will differ by at least one amino acid, and may differ by at least two but not more than about ten amino acids.
  • the sequence changes may be substitutions, insertions or deletions.
  • Conservative amino acid substitutions typically include substitutions within the following groups: (glycine, alanine); (valine, isoleucine, leucine); (aspartic acid, glutamic acid); (asparagine, glutamine); (serine, fhreonine); (lysine, arginine); or (phenylalanine, tyrosine).
  • Modifications of interest that may or may not alter the primary amino acid sequence include chemical derivatization of polypeptides, e.g., acetylation, or carboxylation; changes in amino acid sequence that introduce or remove a glycosylation site; changes in amino acid sequence that make the protein susceptible to PEGylation; and the like. Also included are modifications of glycosylation, e.g. those made by modifying the ' glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g. by exposing the polypeptide to enzymes that affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes. Also embraced are sequences that have phosphorylated amino acid residues, e.g. phosphotyrosine, phosphoserine, or phosphothreonine.
  • modifications of glycosylation e.g. those made by modifying the ' glycosylation patterns of a polypeptide during its synthesis and processing or in
  • the IFN- ⁇ formulation may comprise an N-bloclced species, wherein the N-terminal amino acid is acylated with an acyl group, such as a formyl group, an acetyl group, a malonyl group, and the like. Also suitable for use is a consensus IFN- ⁇ .
  • IFN- ⁇ polypeptides can be produced by any known method. DNA sequences encoding IFN- ⁇ may be synthesized using standard methods. In many embodiments, IFN- ⁇ polypeptides are the products of expression of manufactured DNA sequences transformed or transfected into bacterial hosts, e.g., E. coli, or in eukaryotic host cells (e.g., yeast; mammalian cells, such as CHO cells; and the like). In these embodiments, the IFN- ⁇ is "recombinant IFN- ⁇ .” Where the host cell is a bacterial host cell, the IFN- ⁇ is modified to comprise an N- terminal methionine.
  • IFN- ⁇ as described herein may comprise one or more modified amino acid residues, e.g., glycosylations, chemical modifications, and the like.
  • modified amino acid residues e.g., glycosylations, chemical modifications, and the like.
  • Type II interferon receptor agonists include any naturally occurring or non-naturally- occurring ligand of a human Type II interferon receptor that binds to and causes signal transduction via the receptor.
  • Type II interferon receptor agonists include interferons, including naturally-occurring interferons, modified interferons, synthetic interferons, pegylated interferons, fusion proteins comprising an interferon and a heterologous protein, shuffled interferons; antibody specific for an interferon receptor; non-peptide chemical agonists; and the like.
  • Type II interferon receptor agonist is IFN-gamma and variants thereof. While the present invention exemplifies use of an IFN-gamma polypeptide, it will be readily apparent that any Type II interferon receptor agonist can be used in a subject method. Interferon-Gamma
  • the nucleic acid sequences encoding IFN-gamma polypeptides may be accessed from public databases, e.g., Genbank, journal publications, and the like. While various mammalian IFN-gamma polypeptides are of interest, for the treatment of human disease, generally the human protein will be used. Human IFN-gamma coding sequence may be found in Genbank, accession numbers X13274; V00543; andNM_000619. The corresponding genomic sequence may be found in Genbank, accession numbers J00219; M37265; and V00536. See, for example. Gray et al. (1982) Nature 295:501 (Genbank X13274); and Rinderknecht et al. (1984) J.B.C 259:6790.
  • IFN- ⁇ lb (Actimmune®; human interferon) is a single-chain polypeptide of 140 amino acids. It is made recombinantly in E.coli and is unglycosylated (Rinderknecht et al. 1984, J Biol. Chem. 259:6790-6797). Recombinant IFN-gamma as discussed in U.S. Patent No. 6,497,871 is also suitable for use herein.
  • the IFN-gamma to be used in the methods of the present invention may be any of natural IFN-gamma, recombinant IFN-gamma and the derivatives thereof so far as they have an IFN- ⁇ activity, particularly human IFN-gamma activity.
  • Human IFN-gamma exhibits the antiviral and anti-proliferative properties characteristic of the interferons, as well as a number of other immunomodulatory activities, as is known in the art.
  • IFN-gamma is based on the sequences as provided above, the production of the protein and proteolytic processing can result in processing variants thereof.
  • the unprocessed sequence provided by Gray et al., supra, consists of 166 amino acids (aa).
  • the recombinant IFN-gamma produced in E. coli was originally believed to be 146 amino acids, (commencing at amino acid 20) it was subsequently found that native human IFN-gamma is cleaved after residue 23, to produce a 143 aa protein, or 144 aa if the terminal methionine is present, as required for expression in bacteria.
  • the mature protein can additionally be cleaved at the C terminus after reside 162 (referring to the Gray et al. sequence), resulting in a protein of 139 amino acids, or 140 amino acids if the initial methionine is present, e.g. if required for bacterial expression.
  • the N-terminal methionine is an artifact encoded by the mRNA translational "start" signal AUG that, in the particular case of E. coli expression is not processed away. In other microbial systems or eukaryotic expression systems, methionine may be removed.
  • IIN-gamma peptides of interest include fragments, and can be variously truncated at the carboxyl terminus relative to the full sequence. Such fragments continue to exhibit the characteristic properties of human gamma interferon, so long as amino acids 24 to about 149 (numbering from the residues of the unprocessed polypeptide) are present. Extraneous sequences can be substituted for the amino acid sequence following amino acid 155 without loss of activity. See, for example, U.S. Patent No. 5,690,925.
  • Native IFN- gamma moieties include molecules variously extending from amino acid residues 24-150; 24-151, 24-152; 24- 153, 24-155; and 24-157. Any of these variants, and other variants known in the art and having IFN- ⁇ activity, may be used in the present methods.
  • the sequence of the IFN- ⁇ polypeptide may be altered in various ways known in the art to generate targeted changes in sequence.
  • a variant polypeptide will usually be substantially similar to the sequences provided herein, i.e., will differ by at least one amino acid, and may differ by at least two but not more than about ten amino acids.
  • the sequence changes may be substitutions, insertions or deletions.
  • Scanning mutations that systematically introduce alanine, or other residues, may be used to determine key amino acids.
  • Specific amino acid substitutions of interest include conservative and non-conservative changes.
  • Conservative amino acid substitutions typically include substitutions within the following groups: (glycine, alanine); (valine, isoleucine, leucine); (aspartic acid, glutamic acid); (asparagine, glutamine); (serine, threonine); (lysine, arginine); or (phenylalanine, tyrosine).
  • Modifications of interest that may or may not alter the primary amino acid sequence include chemical derivatization of polypeptides, e.g., acetylation, or carboxylation; changes in amino acid sequence that introduce or remove a glycosylation site; changes in amino acid sequence that make the protein susceptible to PEGylation; and the like.
  • IFN-gamma may be modified with one or more polyethylene glycol moieties (PEGylated).
  • the invention contemplates the use of IFN-gamma variants with one or more non-naturally occurring glycosylation and/or pegylation sites that are engineered to provide glycosyl- and/or PEG-derivatized polypeptides with reduced serum clearance, such as the IFN-gamma polypeptide variants described in International Patent Publication No. WO 01/36001.
  • modifications of glycosylation e.g., those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g., by exposing the polypeptide to enzymes that affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes.
  • sequences that have phosphorylated amino acid residues e.g., phosphotyrosine, phosphoserine, or phosphothreonine.
  • polypeptides that have been modified using ordinary chemical techniques so as to improve their resistance to proteolytic degradation, to optimize solubility properties, or to render them more suitable as a therapeutic agent.
  • the backbone of the peptide may be cyclized to enhance stability (see, for example, Friedler et al. 2000, J Biol. Chem. 275:23783-23789).
  • Analogs may be used that include residues other than naturally occurring L-amino acids, e.g., D-amino acids or non-naturally occurring synthetic amino acids.
  • the protein may be pegylated to enhance stability.
  • the polypeptides may be prepared by in vitro synthesis, using conventional methods as known in the art, by recombinant methods, or may be isolated from cells induced or naturally producing the protein. The particular sequence and the manner of preparation will be determined by convenience, economics, purity required, and the like. If desired, various groups may be introduced into the polypeptide during synthesis or during expression, which allow for linking to other molecules or to a surface. Thus cysteines can be used to make thioethers, histidines for linking to a metal ion complex, carboxyl groups for forming amides or esters, amino groups for forming amides, and the like.
  • the polypeptides may also be isolated and purified in accordance with conventional methods of recombinant synthesis.
  • a lysate may be prepared of the expression host and the lysate purified using HPLC, exclusion chromatography, gel electrophoresis, affinity chromatography, or other purification technique.
  • the compositions which are used will comprise at least 20% by weight of the desired product, more usually at least about 15% by weight, preferably at least about 95%) by weight, and for therapeutic purposes, usually at least about 99.5%> by weight, in relation to contaminants related to the method of preparation of the product and its purification. Usually, the percentages will be based upon total protein.
  • Suitable TNF- ⁇ antagonists for use herein include agents that decrease the level of TNF- ⁇ synthesis, agents that block or inhibit the binding of TNF- ⁇ to a TNF- ⁇ receptor (TNFR), and agents that block or inhibit TNFR-mediated signal transduction.
  • TNFR TNF- ⁇ receptor
  • every reference to a "TNF- ⁇ antagonist” or “T ⁇ F antagonist” herein will be understood to mean a TTSfF- ⁇ antagonist other than pirfenidone or a pirfenidone analog.
  • TNF receptor polypeptide and "T ⁇ FR polypeptide” refer to polypeptides derived from TNFR (from any species) which are capable of binding TNF.
  • T ⁇ FR Two distinct cell-surface TNFRs have described: Type II T ⁇ FR (or p75 TNFR or T ⁇ FRII) and Type I T ⁇ FR (or p55 T ⁇ FR or T ⁇ FRI).
  • Type II T ⁇ FR or p75 TNFR or T ⁇ FRII
  • Type I T ⁇ FR or p55 T ⁇ FR or T ⁇ FRI
  • the mature full-length human p75 T ⁇ FR is a glycoprotein having a molecular weight of about 75-80 kilodaltons (kD).
  • the mature full- length human p55 TNFR is a glycoprotein having a molecular weight of about 55-60 kD.
  • Exemplary TNFR polypeptides are derived from TNFR Type I and/or TNFR type II.
  • Soluble TNFR includes p75 TNFR polypeptide; fusions of p75 TNFR with heterologous fusion partners, e.g., the Fc portion of an immunoglobulin.
  • TNFR polypeptide may be an intact TNFR or a suitable fragment of T ⁇ FR.
  • U.S. Pat. No. 5,605,690 provides examples of TNFR polypeptides, including soluble T ⁇ FR polypeptides, appropriate for use in the present invention.
  • the T ⁇ FR polypeptide comprises an extracellular domain of TNFR.
  • the TNFR polypeptide is a fusion polypeptide comprising an extracellular domain of TNFR linked to a constant domain of an immunoglobulin molecule, hi other embodiments, the T ⁇ FR polypeptide is a fusion polypeptide comprising an extracellular domain of the p75 T ⁇ FR linked to a constant domain of an IgGl molecule.
  • an Ig used for fusion proteins is human, e.g., human IgGl .
  • T ⁇ FR polypeptides may be used in the present invention.
  • Multivalent forms of TNFR polypeptides possess more than one TNF binding site.
  • the T ⁇ FR is a bivalent, or dimeric, form of TNFR.
  • a chimeric antibody polypeptide with TNFR extracellular domains substituted for the variable domains of either or both of the immunoglobulin heavy or light chains would provide a TNFR polypeptide for the present invention.
  • T ⁇ FR antibody polypeptide
  • a subject method involves administration of an effective amount of the soluble TNFR ENBREL®.
  • ENBREL® is a dimeric fusion protein consisting of the extracellular ligand-binding portion of the human 75 kilodalton (pi 5) TNFR linked to the Fc portion of human IgGl.
  • the Fc component of ENBREL® contains the CH2 domain, the CH3 domain and hinge region, but not the CHI domain of IgGl.
  • ENBREL® is produced in a Chinese hamster ovary (CHO) mammalian cell expression system. It consists of 934 amino acids and has an apparent molecular weight of approximately 150 kilodaltons. Smith et al.
  • Monoclonal antibodies that bind TNF- ⁇ .
  • Monoclonal antibodies include "humanized" mouse monoclonal antibodies; chimeric antibodies; monoclonal antibodies that are at least about 80%), at least about 90%, at least about 95%, or 100% human in amino acid sequence; and the like. See, e.g., WO 90/10077; WO 90/04036; and WO 92/02190.
  • Suitable monoclonal antibodies include antibody fragments, such as Fv, F(ab') 2 and Fab; synthetic antibodies; artificial antibodies; phage display antibodies; and the like.
  • REMICADE® is a chimeric monoclonal anti-TNF- ⁇ antibody that includes about 25% mouse amino acid sequence and about 75% human amino acid sequence.
  • REMICADE® comprises a variable region of a mouse monoclonal anti-TNF- ⁇ antibody fused to the constant region of a human IgGl .
  • HUMIRATM is a human, full-length IgGl monoclonal antibody that was identified using phage display technology. Piascik (2003) J Am. Pharm. Assoc. 43:327-328.
  • SAPK inhibitors are known in the art, and include, but are not limited to 2-alkyl imidazoles disclosed in U.S. Patent No. 6,548,520; 1,4,5-substituted imidazole compounds disclosed in U.S. Patent No. 6,489,325; 1,4,5-substituted imidazole compounds disclosed in U.S. Patent No. 6,569,871; heteroaryl aminophenyl ketone compounds disclosed in Published U.S. Patent Application No. 2003/0073832; pyridyl imidazole compounds disclosed in U.S. Patent No.
  • a stress-activated protein kinase is a member of a family of mitogen- activated protein kinases which are activated in response to stress stimuli.
  • SAPK include, but are not limited to, p38 (Lee et al. (1994) Nature 372:739) and c-jun N-terminal kinase (JNK).
  • TNF antagonist activity may be assessed with a cell-based competitive binding assay.
  • radiolabeled TNF is mixed with serially diluted TNF antagonist and cells expressing cell membrane bound TNFR. Portions of the suspension are centrifuged to separate free and bound TNF and the amount of radioactivity in the free and bound fractions determined. TNF antagonist activity is assessed by inhibition of TNF binding to the cells in the presence of the TNF antagonist.
  • TNF antagonists may be analyzed for the ability to neutralize TNF activity in vitro in a bioassay using cells susceptible to the cytotoxic activity of TNF as target cells.
  • target cells cultured with TNF, are treated with varying amounts of TNF antagonist and subsequently are examined for cytolysis.
  • TNF antagonist activity is assessed by a decrease in TNF-induced target cell cytolysis in the presence of the TNF antagonist.
  • Pirfenidone and Analogs Thereof Pirfenidone (5 -methyl- l-phenyl-2-(lH)-pyridone) and pirfenidone analogs are used in certain combination therapies of the invention. Pirfenidone
  • Ri carbocyclic (saturated and unsaturated), heterocyclic (saturated or unsaturated), alkyls (saturated and unsaturated). Examples include phenyl, benzyl, pyrimidyl, naphthyl, indolyl, pyrrolyl, furyl, thienyl, imidazolyl, cyclohexyl, piperidyl, pyrrolidyl, morpholinyl, cyclohexenyl, butadienyl, and the like.
  • Ri can further include substitutions on the carbocyclic or heterocyclic moieties with substituents such as halogen, nitro, amino, hydroxyl, alkoxy, carboxyl, cyano, thio, alkyl, aryl, heteroalkyl, heteroaryl and combinations thereof, for example, 4-nitrophenyl, 3-chloro ⁇ henyl, 2,5-dinitrophenyl, 4-methoxyphenyl, 5-methyl-pyrrolyl, 2, 5-dichlorocyclohexyl, guanidinyl- cyclohexenyl and the like.
  • substituents such as halogen, nitro, amino, hydroxyl, alkoxy, carboxyl, cyano, thio, alkyl, aryl, heteroalkyl, heteroaryl and combinations thereof, for example, 4-nitrophenyl, 3-chloro ⁇ henyl, 2,5-dinitrophenyl, 4-methoxyphenyl, 5-methyl-pyrrolyl, 2, 5-dich
  • R 2 alkyl, carbocylic, aryl, heterocyclic. Examples include: methyl, ethyl, propyl, isopropyl, phenyl, 4-nitrophenyl, thienyl and the like.
  • X may be any number (from 1 to 3) of substituents on the carbocyclic or heterocyclic ring.
  • the substituents can be the same or different.
  • Substituents can include hydrogen, alkyl, heteroalkyl, aryl, heteroaryl, halo, nitro, carboxyl, hydroxyl, cyano, amino, thio, alkylamino, haloaryl and the like.
  • the substituents may be optionally further substituted with 1-3 substituents from the group consisting of alkyl, aryl, nitro, alkoxy, hydroxyl and halo groups.
  • Examples include: methyl, 2,3-dimethyl, phenyl, p-tolyl, 4-chlorophenyl, 4-nitrophenyl, 2,5-dichlorophenyl, furyl, thienyl and the like. [00154] Specific Examples include those shown in Table 1 :
  • a subject combination therapy further includes administering a side effect management agent that reduces a side effect of a therapeutic agent, in an amount effective to reduce at least one side effect.
  • Side effect management agents include palliative agents, and other agents for the treatment, reduction, or avoidance of a side effect caused by any therapeutic agent.
  • a subject therapy with pirfenidone or pirfenidone analog further includes administering a side effect management agent in an amount effective to reduce at least one side effect of the pirfenidone or pirfenidone analog therapy.
  • Side effects of pirfenidone or pirfenidone analog treatment include gastrointestinal disturbances and discomfort. Gastrointestinal disturbances include nausea, diarrhea, gastrointestinal cramping, and the like.
  • an effective amount of a side effect management agent reduces a side effect induced by treatment with a pirfenidone or pirfenidone analog by at least about 10%, at least about 20%, at least about 30%), at least about 40%, at least about 50%, at least about 60%, or more, compared to the rate of occurrence or the degree or extent of the side effect when the pirfenidone or pirfenidone analog therapy is administered without the side effect management agent.
  • a subject therapy with a Type II interferon receptor agonist further includes administering a side effect management agent in an amount effective to reduce at least one side effect of the Type II interferon receptor agonist therapy.
  • Side effects of Type II interferon receptor agonist treatment include, but are not limited to, fever, malaise, tachycardia, chills, headache, arthralgia, myalgia, myelosuppression, suicide ideation, platelet suppression, neutropenia, lymphocytopenia, erythrocytopenia (anemia), and anorexia.
  • an effective amount of a side effect management agent reduces a side effect induced by treatment with a Type II interferon receptor agonist by at least about 10%), at least about 20%>, at least about 30%>, at least about 40%, at least about 50%, at least about 60%>, or more, compared to the rate of occurrence or the degree or extent of the side effect when the Type II interferon receptor agonist therapy is administered without the side effect management agent.
  • the body temperature of an individual treated with the Type II interferon receptor agonist therapy and side effect management agent according to the instant invention is reduced by at least 0.5 degree Fahrenheit, and in some embodiments is within the normal range, e.g., at or near 98.6 °F.
  • Suitable side effect management agents include agents that are effective in pain management; agents that ameliorate gastrointestinal discomfort; analgesics, anti- inflammatories, antipsychotics, antineurotics, anxiolytics, hematopoietic agents, and agents that reduce gastrointestinal discomfort.
  • the invention contemplates the use of any compound for palliative care of patients suffering from pain or any other side effect in the course of treatment with a subject monotherapy or combination therapy.
  • Exemplary side effect management agents include acetaminophen, ibuprofen, and other NSAIDs, H2 blockers, hematopoietic agents, and antacids.
  • H2 blockers that are suitable for use as a side effect management agent in a subject therapy include, but are not limited to, Cimetidine (e.g., Tagamet, Peptol, Nu-cimet, apo-cimetidine, non-cimetidine); Ranitidine (e.g., Zantac, Nu-ranit, Novo-randine, and apo-ranitidine); and Famotidine (Pepcid, Apo-Famotidine, and No vo-Famotidine) .
  • Cimetidine e.g., Tagamet, Peptol, Nu-cimet, apo-cimetidine, non-cimetidine
  • Ranitidine e.g., Zantac, Nu-ranit, Novo-randine, and apo-ranitidine
  • Famotidine Pepcid, Apo-Famotidine, and No vo-Famotidine
  • Suitable antacids include, but are not limited to, aluminum and magnesium hydroxide (Maalox®, Mylanta®); aluminum carbonate gel (Basajel®); aluminum hydroxide (Amphojel®, AlternaGEL®); calcium carbonate (Turns®, Titralac®); magnesium hydroxide; and sodium bicarbonate.
  • Suitable non-steroidal anti-inflammatory drugs include, but are not limited to, acetaminophen, salicylate, acetyl-salicylic acid (aspirin, diflunisal), ibuprofen, Motrin, Naprosyn, Nalfon, and Trilisate, indomethacin, glucametacme, acemetacin, sulindac, naproxen, piroxicam, diclofenac, benoxaprofen, ketoprofen, oxaprozin, etodolac, Icetorolac tromethamine, ketorolac, nabumetone, and the like, and mixtures of two or more of the foregoing.
  • Suitable hematopoietic agents include agents that prevent or restore depressed blood cell populations, including, but not limited to, erythropoietins, such as EPOGENTM epoetin- alfa, granulocyte colony stimulating factors (G-CSFs), such as NEUPOGENTM filgrastim, granulocyte-macrophage colony stimulating factors (GM-CSFs), thrombopoietins, etc. DOSAGES, FORMULATIONS, AND ROUTES OF ADMINISTRATION
  • a therapeutic agent e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist
  • pirfenidone or pirfenidone analog which is administered in a subject monotherapy or combination therapy is administered to individuals in a formulation with a pharmaceutically acceptable excipient(s).
  • a pharmaceutically acceptable excipient(s) are known in the art and need not be discussed in detail herein. Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, A. Gennaro (2000) "Remington: The Science and Practice of Pharmacy," 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H.C.
  • compositions such as vehicles, adjuvants, carriers or diluents
  • pharmaceutically acceptable auxiliary substances such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
  • an active agent e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist
  • an active agent e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist
  • the agents can be incorporated into a variety of formulations for therapeutic administration.
  • an active agent can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.
  • an active agent e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist
  • an active agent e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist
  • oral, buccal, rectal, parenteral, intraperitoneal, intradermal, subcutaneous, intramuscular, transdermal, intratracheal, etc. administration.
  • two different routes of administration are used.
  • Subcutaneous administration of a therapeutic agent can be accomplished using standard methods and devices, e.g., needle and syringe, a subcutaneous injection port delivery system, and the like. See, e.g., U.S. Patent Nos. 3,547,119; 4,755,173; 4,531,937; 4,311,137; and 6,017,328.
  • a combination of a subcutaneous injection port and a device for administration of an agent to a patient through the port is referred to herein as "a subcutaneous injection port delivery system.”
  • subcutaneous administration is achieved by a combination of devices, e.g., bolus delivery by needle and syringe, followed by delivery using a continuous delivery system.
  • a therapeutic agent is delivered by a continuous delivery system.
  • continuous delivery system is used interchangeably herein with “controlled delivery system” and encompasses continuous (e.g., controlled) delivery devices (e.g., pumps) in combination with catheters, injection devices, and the like, a wide variety of which are known in the art.
  • Mechanical or electromechanical infusion pumps can also be suitable for use with the present invention.
  • Examples of such devices include those described in, for example, U.S. Pat. Nos. 4,692,147; 4,360,019; 4,487,603; 4,360,019; 4,725,852; 5,820,589; 5,643,207; 6,198,966; and the like.
  • the present methods of drug delivery can be accomplished using any of a variety of refillable, pump systems. Pumps provide consistent, controlled release over time.
  • the therapeutic agent is in a liquid formulation in a drug-impermeable reservoir, and is delivered in a continuous fashion to the individual.
  • the drug delivery system is an at least partially implantable device.
  • the implantable device can be implanted at any suitable implantation site using methods and devices well known in the art.
  • An implantation site is a site within the body of a subject at which a drug delivery device is introduced and positioned.
  • Implantation sites include, but are not necessarily limited to a subdermal, subcutaneous, intramuscular, or other suitable site within a subject's body. Subcutaneous implantation sites are generally preferred because of convenience in implantation and removal of the drug delivery device.
  • Drug release devices suitable for use in the invention may be based on any of a variety of modes of operation.
  • the drug release device can be based upon a diffusive system, a convective system, or an erodible system (e.g., an erosion-based system).
  • the drug release device can be an electrochemical pump, osmotic pump, an electroosmotic pump, a vapor pressure pump, or osmotic bursting matrix, e.g., where the drug is incorporated into a polymer and the polymer provides for release of drug formulation concomitant with degradation of a drug-impregnated polymeric material (e.g., a biodegradable, drug-impregnated polymeric material).
  • the drug release device is based upon an electrodiffusion system, an electrolytic pump, an effervescent pump, a piezoelectric pump, a hydrolytic system, etc.
  • Drug release devices based upon a mechanical or electromechanical infusion pump can also be suitable for use with the present invention. Examples of such devices include those described in, for example, U.S. Pat. Nos. 4,692,147; 4,360,019; 4,487,603; 4,360,019; 4,725,852, and the like.
  • the present methods of drug delivery can be accomplished using any of a variety of refillable, non-exchangeable pump systems. Pumps and other convective systems are generally preferred due to their generally more consistent, controlled release over time. Osmotic pumps are used in some embodiments, due to their combined advantages of more consistent controlled release and relatively small size (see, e.g., PCT published application no. WO 97/27840 and U.S.
  • Exemplary osmotically-driven devices suitable for use in the invention include, but are not necessarily limited to, those described in U.S. Pat. Nos. 3,760,984; 3,845,770; 3,916,899; 3,923,426; 3,987,790; 3,995,631; 3,916,899; 4,016,880; 4,036,228; 4,111,202; 4,111,203; 4,203,440; 4,203,442; 4,210,139; 4,327,725; 4,627,850; 4,865,845; 5,057,318; 5,059,423; 5,112,614; 5,137,727; 5,234,692; 5,234,693; 5,728,396; and the like.
  • the drug delivery device is an implantable device.
  • the drug delivery device can be implanted at any suitable implantation site using methods and devices well known in the art.
  • an implantation site is a site within the body of a subject at which a drug delivery device is introduced and positioned. Implantation sites include, but are not necessarily limited to a subdermal, subcutaneous, intramuscular, or other suitable site within a subject's body.
  • a therapeutic agent is delivered using an implantable drug delivery system, e.g., a system that is programmable to provide for administration of a an active agent (e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist).
  • a an active agent e.g., pirfenidone or pirfenidone analog
  • a Type I interferon receptor agonist e.g., pirfenidone or pirfenidone analog
  • Type I interferon receptor agonist e.g., pirfenidone or pirfenidone analog
  • Type I interferon receptor agonist e.g., pirfenidone or pirfenidone analog
  • a Type II interferon receptor agonist e.g., pirfenidone analog
  • TNF antagonist e.g.,
  • a further exemplary device that can be adapted for the present invention is the Syncl romed infusion pump (Medtronic).
  • an active agent e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist
  • an active agent may be administered in the form of its pharmaceutically acceptable salts, or the active agent may also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
  • the following methods and excipients are merely exemplary and are in no way limiting.
  • an active agent can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
  • conventional additives such as lactose, mannitol, corn starch or potato starch
  • binders such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins
  • disintegrators such as corn starch, potato starch or sodium carboxymethylcellulose
  • lubricants such as talc or magnesium stearate
  • the agents can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • an aqueous or nonaqueous solvent such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol
  • solubilizers isotonic agents
  • suspending agents emulsifying agents
  • stabilizers and preservatives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
  • the agents can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases.
  • bases such as emulsifying bases or water-soluble bases.
  • the compounds of the present invention can be administered rectally via a suppository.
  • the suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at room temperature.
  • Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more active agents.
  • unit dosage forms for injection or intravenous administration may comprise the active agent(s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of compounds of the present invention calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • the specifications for an active agent depend on the particular agent employed and the effect to be achieved, and the pharmacodynamics associated with the agent in the host.
  • the invention provides embodiments in which the therapeutic agent administered to the patient by a controlled drug delivery device.
  • the therapeutic agent is delivered to the patient substantially continuously or continuously by the controlled drug delivery device.
  • an implantable infusion pump is used to deliver the therapeutic agent to the patient substantially continuously or continuously by subcutaneous infusion.
  • a therapeutic agent is administered to the patient so as to achieve and maintain a desired average daily serum concentration of the therapeutic agent at a substantially steady state for the duration of the therapy.
  • an implantable infusion pump is used to deliver the therapeutic agent to the patient by subcutaneous infusion so as to achieve and maintain a desired average daily serum concentration of the therapeutic agent at a substantially steady state for the duration of the therapy.
  • Effective dosages of pirfenidone or a specific pirfenidone analog include a weight- based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally.
  • Other doses and formulations of pirfenidone and specific pirfenidone analogs suitable for use in the treatment of fibrotic diseases are described in U.S. Pat. Nos., 5,310,562; 5,518,729; 5,716,632; and 6,090,822.
  • a pirfenidone or pirfenidone analog is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time.
  • the pirfenidone or pirfenidone analog can be administered fid, bid, qd, qod, biw, tiw, qw, qow, three times per month, once monthly, substantially continuously, or continuously.
  • a pirfenidone or pirfenidone analog is administered once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (bid), or three times a day (tid), substantially continuously, or continuously, over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.
  • a pirfenidone or pirfenidone analog is administered in combination therapy with at least a second therapeutic agent.
  • a second therapeutic agent is administered throughout the entire course of pirfenidone/pirfenidone analog treatment.
  • a second therapeutic agent is administered less than the entire course of pirfenidone/pirfenidone analog treatment, e.g., only during the first phase of pirfenidone/pirfenidone analog treatment, only during the second phase of pirfenidone/pirfenidone analog treatment, or some other portion of the pirfenidone/pirfenidone analog treatment regimen.
  • a subject combination therapy involves co-administering pirfenidone or a pirfenidone analog and a Type II interferon receptor agonist.
  • the Type II interferon receptor agonist is an IFN- ⁇ .
  • Effective dosages of IFN- ⁇ can range from about 0.5 ⁇ g/m 2 to about 500 ⁇ g/m 2 , usually from about 1.5 ⁇ g/m 2 to 200 ⁇ g/m 2 , depending on the size of the patient. This activity is based on 10 6 international units (U) per 50 ⁇ g of protein. IFN- ⁇ can be administered daily, every other day, three times a week, or substantially continuously or continuously.
  • IFN- ⁇ is administered to an individual in a unit dosage form of from about 25 ⁇ g to about 500 ⁇ g, from about 50 ⁇ g to about 400 ⁇ g, or from about 100 ⁇ g to about 300 ⁇ g. In particular embodiments of interest, the dose is about 200 ⁇ g IFN- ⁇ . In many embodiments of interest, IFN- ⁇ lb is administered.
  • the amount of IF ⁇ - ⁇ per body weight (assuming a range of body weights of from about 45 kg to about 135 kg) is in the range of from about 4.4 ⁇ g IFN- ⁇ per kg body weight to about 1.48 ⁇ g IFN- ⁇ per kg body weight.
  • the body surface area of subject individuals generally ranges from about 1.33 m 2 to about 2.50 m 2 .
  • an IFN- ⁇ dosage ranges from about 150 ⁇ g/m 2 to about 20 ⁇ g/m 2 .
  • an IFN- ⁇ dosage ranges from about 20 ⁇ g/m 2 to about 30 ⁇ g/m 2 , from about 30 ⁇ g/m 2 to about 40 ⁇ g/m 2 , from about 40 ⁇ g/m 2 to about 50 ⁇ g/m 2 , from about 50 ⁇ g/m 2 to about 60 ⁇ g/m 2 , from about 60 ⁇ g/m 2 to about 70 ⁇ g/m 2 , from about 70 ⁇ g/m 2 to about 80 ⁇ g/m 2 , from about 80 ⁇ g/m 2 to about 90 ⁇ g/m 2 , from about 90 ⁇ g/m 2 to about 100 ⁇ g/m 2 , from about 100 ⁇ g/m 2 to about 110 ⁇ g/m 2 , from about 110 ⁇ g/m 2 to about 120 ⁇ g/m 2 , from about 120 ⁇ g/m 2 to about 130 ⁇ g/m 2 , from about 130 ⁇ g/m 2 to about 140 ⁇ g/m 2 , or from about 140 ⁇
  • the dosage groups range from about 25 ⁇ g/m 2 to about 100 ⁇ g/m 2 . In other embodiments, the dosage groups range from about 25 ⁇ g/m to about 50 ⁇ g/m .
  • IFN- ⁇ is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time.
  • IFN- ⁇ can be administered tid, bid, qd, qod, biw, tiw, qw, qow, three times per month, once monthly, substantially continuously, or continuously.
  • IFN- ⁇ is administered once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (bid), or three times a day (tid), substantially continuously, or continuously, over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.
  • a subject combination therapy involves administering a Type I interferon receptor agonist.
  • the Type I interferon receptor agonist is an IFN- ⁇ . Effective dosages of an IFN- ⁇ range from about 3 ⁇ g to about 27 ⁇ g, from about 3 MU to about 10 MU, from about 90 ⁇ g to about 180 ⁇ g, or from about 18 ⁇ g to about 90 ⁇ g.
  • Effective dosages of Infergen® consensus IFN- ⁇ include about 3 ⁇ g, about 6 ⁇ g, about 9 ⁇ g, about 12 ⁇ g, about 15 ⁇ g, about 18 ⁇ g, about 21 ⁇ g, about 24 ⁇ g, about 27 ⁇ g, or about 30 ⁇ g, of drug per dose.
  • Effective dosages of IFN- ⁇ 2a and IFN- ⁇ 2b range from 3 million Units (MU) to 10 MU per dose.
  • Effective dosages of PEGASYS®PEGylated IFN- ⁇ 2a contain an amount of about 90 ⁇ g to 270 ⁇ g, or about 180 ⁇ g, of drug per dose.
  • Effective dosages of PEG-INTRON®PEGylated IF ⁇ - ⁇ 2b contain an amount of about 0.5 ⁇ g to 3.0 ⁇ g of drug per kg of body weight per dose.
  • Effective dosages of PEGylated consensus interferon (PEG- CIFN) contain an amount of about 18 ⁇ g to about 90 ⁇ g, or from about 27 ⁇ g to about 60 ⁇ g, or about 45 ⁇ g, of CIFN amino acid weight per dose of PEG-CIFN.
  • Effective dosages of monoPEG (30 kD, linear)-ylated CIFN contain an amount of about 45 ⁇ g to about 270 ⁇ g, or about 60 ⁇ g to about 180 ⁇ g, or about 90 ⁇ g to about 120 ⁇ g, of drug per dose.
  • IFN- ⁇ can be administered daily, every other day, once a week, three times a week, every other week, three times per month, once monthly, substantially continuously or continuously.
  • a subject combination therapy involves administering a TNF- ⁇ antagonist.
  • Effective dosages of a TNF- ⁇ antagonist range from 0.1 ⁇ g to 40 mg per dose, e.g., from about 0.1 ⁇ g to about 0.5 ⁇ g per dose, from about 0.5 ⁇ g to about 1.0 ⁇ g per dose, from about 1.0 ⁇ g per dose to about 5.0 ⁇ g per dose, from about 5.0 ⁇ g to about 10 ⁇ g per dose, from about 10 ⁇ g to about 20 ⁇ g per dose, from about 20 ⁇ g per dose to about 30 ⁇ g per dose, from about 30 ⁇ g per dose to about 40 ⁇ g per dose, from about 40 ⁇ g per dose to about 50 ⁇ g per dose, from about 50 ⁇ g per dose to about 60 ⁇ g per dose, from about 60 ⁇ g per dose to about 70 ⁇ g per dose, from about 70 ⁇ g to about 80 ⁇ g per dose, from about 80 ⁇ g per dose to about 100 ⁇ g per dose, from about 100 ⁇ g to about 150 ⁇ g per dose, from about 150 ⁇ g to about
  • the TNF- ⁇ antagonist is ENBREL® etanercept.
  • Effective dosages of etanercept range from about 0.1 ⁇ g to about 40 mg per dose, from about 0.1 ⁇ g to about 1 ⁇ g per dose, from about 1 ⁇ g to about 10 ⁇ g per dose, from about 10 ⁇ g to about 100 ⁇ g per dose, from about 100 ⁇ g to about 1 mg per dose, from about 1 mg to about 5 mg per dose, from about 5 mg to about 10 mg, from about 10 mg to about 15 mg per dose, from about 15 mg to about 20 mg per dose, from about 20 mg to about 25 mg per dose, from about 25 mg to about 30 mg per dose, from about 30 mg to about 35 mg per dose, or from about 35 mg to about 40 mg per dose.
  • effective dosages of a TNF- ⁇ antagonist are expressed as mg/kg body weight.
  • effective dosages of a TNF- ⁇ antagonist are from about 0.1 mg/kg body weight to about 10 mg/kg body weight, e.g., from about 0.1 mg/kg body weight to about 0.5 mg/kg body weight, from about 0.5 mg/kg body weight to about 1.0 mg/kg body weight, from about 1.0 mg/kg body weight to about 2.5 mg/kg body weight, from about 2.5 mg/kg body weight to about 5.0 mg/kg body weight, from about 5.0 mg/kg body weight to about 7.5 mg/kg body weight, or from about 7.5 mg/kg body weight to about 10 mg/kg body weight.
  • the TNF- ⁇ antagonist is REMICADE®.
  • Effective dosages of REMICADE® range from about 0.1 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 0.5 mg/kg, from about 0.5 mg/kg to about 1.0 mg/kg, from about 1.0 mg/kg to about 1.5 mg/kg, from about 1.5 mg/kg to about 2.0 mg/kg, from about 2.0 mg/kg to about 2.5 mg/kg, from about 2.5 mg/kg to about 3.0 mg/kg, from about 3.0 mg/kg to about 3.5 mg/kg, from about 3.5 mg/kg to about 4.0 mg/kg, from about 4.0 mg/kg to about 4.5 mg/kg, from about 4.5 mg/kg to about 5.0 mg/kg, from about 5.0 mg/kg to about 7.5 mg/kg, or from about 7.5 mg/kg to about 10 mg/kg per dose.
  • the TNF- ⁇ antagonist is HUMIRATM.
  • Effective dosages of HUMIRATM range from about 0.1 ⁇ g to about 35 mg, from about- 0.1 ⁇ g to about 1 ⁇ g, from about 1 ⁇ g to about 10 ⁇ g, from about 10 ⁇ g to about 100 ⁇ g, from about 100 ⁇ g to about 1 mg, from about 1 mg to about 5 mg, from about 5 mg to about 10 mg, from about 10 mg to about 15 mg, from about 15 mg to about 20 mg, from about 20 mg to about 25 mg, from about 25 mg to about 30 mg, from about 30 mg to about 35 mg, or from about 35 mg to about 40 mg per dose.
  • a TNF- ⁇ antagonist is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time.
  • the TNF- ⁇ antagonist can be administered tid, bid, qd, qod, biw, tiw, qw, qow, three times per month, once monthly, once bimonthly, substantially continuously, or continuously.
  • a TNF- ⁇ antagonist is administered once bimonthly, once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (bid), or three times a day (tid), substantially continuously, or continuously, over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.
  • dose levels can vary as a function of the specific compounds, the severity of the symptoms and the susceptibility of the subject to side effects.
  • Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means. A preferred means is to measure the physiological potency of a given compound.
  • the present invention provides pirfenidone or pirfenidone analog monotherapy for the treatment of alcoholic hepatitis. In another aspect, the present invention provides pirfenidone or pirfenidone analog combination therapy for the treatment of alcoholic hepatitis. In another aspect, the present invention provides pirfenidone or pirfenidone analog monotherapy therapy for the treatment of ALD. In another aspect, the present invention provides combination therapy for the treatment of ALD. Pirfenidone monotherapy for treating non-fibrotic alcoholic hepatitis
  • the present invention provides pirfenidone or pirfenidone analog monotherapy for the treatment of non-fibrotic alcoholic hepatitis.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd, optionally in two or more divided doses per day, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog of about 1200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog of about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the present invention provides pirfenidone or pirfenidone analog combination therapy for the treatment of non-fibrotic alcoholic hepatitis.
  • the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog and a Type II interferon receptor agonist in the treatment of non-fibrotic alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; and an effective amount of
  • the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog and a TNF- ⁇ antagonist in the treatment of non-fibrotic alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; and an effective amount of a TNF- ⁇ antagonist.
  • the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog, a Type II interferon receptor agonist and a TNF- ⁇ antagonist in the treatment of non-fibrotic alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; an effective amount of a Type II interferon receptor agonist; and an effective amount of a TNF- ⁇ antagonist.
  • the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog, a Type II interferon receptor agonist, a Type I interferon receptor agonist and a TNF- ⁇ antagonist in the treatment of non-fibrotic alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; an effective amount of a Type II interferon receptor agonist; an effective amount of a Type I interferon receptor agonist; and an effective amount
  • the effective amount of a Type II interferon receptor agonist referred to in any of the foregoing methods of pirfenidone combination therapy for treatment of non-fibrotic alcoholic hepatitis can be selected from a fixed dosage of IFN- ⁇ in the range from about 25 ⁇ g to about 500 ⁇ g, from about 50 ⁇ g to about 400 ⁇ g, or from about 100 ⁇ g to about 300 ⁇ g, or about 200 ⁇ g, of IFN- ⁇ per dose subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously for the duration of pirfenidone therapy.
  • the effective amount of a Type I interferon receptor agonist referred to in any of the foregoing methods of pirfenidone combination therapy for treatment of non-fibrotic alcoholic hepatitis can be selected from: (1) a fixed dosage of Infergen® consensus IFN- ⁇ of about 3 ⁇ g, about 6 ⁇ g, about 9 ⁇ g, about 12 ⁇ g, about 15 ⁇ g, about 18 ⁇ g, about 21 ⁇ g, about 24 ⁇ g, about 27 ⁇ g, or about 30 ⁇ g, of drug per dose subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously for the duration of pirfenidone therapy; (2) a fixed dosage of IFN- ⁇ 2a, 2b or 2c in the range from about 3 million Units (MU) to 10 MU of drug per dose subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously for the duration of pir
  • the effective amount of a TNF- ⁇ antagonist referred to in any of the foregoing methods of pirfenidone combination therapy for treatment of non-fibrotic alcoholic hepatitis can be selected from: (1) a fixed dosage of ENBREL® in the range of about 10 mg to about 40 mg, or about 25 mg, of drug per dose subcutaneously biw for the duration of pirfenidone therapy; (2) a weight-based dosage of REMICADE® in the range of about 3 mg to about 10 mg of drug per kilogram of body weight intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks for the duration of pirfenidone therapy; or (3) a fixed dosage of HUMIRATM of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks for the duration of pirfenidone therapy.
  • the present invention provides pirfenidone or pirfenidone analog monotherapy therapy for the treatment of ALD.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd, optionally in two or more divided doses per day, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
  • Pirfenidone combination therapy (other than IFN- ⁇ ) for the treatment of ALD
  • the present invention provides combination therapy for the treatment of ALD, comprising administering pirfenidone or a pirfenidone and at least a second therapeutic agent other than IFN- ⁇ in combined amounts effective to treat ALD.
  • the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; and an effective amount of a TNF- ⁇ antagonist.
  • the effective amount of TNF- ⁇ antagonist can be selected from: (1) a fixed dosage of ENBREL® in the range of about 10 mg to about 40 mg, or about 25 mg, of drug per dose subcutaneously biw for the duration of pirfenidone therapy; (2) a weight-based dosage of REMICADE® in the range of about 3 mg to about 10 mg of drug per kilogram of body weight intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks for the duration of pirfenidone therapy; or (3) a fixed dosage of HUMIRATM of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks for the duration of pirfenidone therapy.
  • the present invention provides combination therapy for the treatment of ALD, comprising administering an effective amount of a Type II interferon receptor agonist, an effective amount of pirfenidone or a pirfenidone analog, and a side effect management agent.
  • the method employs a Type II interferon receptor agonist that is IFN-gamma (IFN- ⁇ ).
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising co-administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a dosage of pirfenidone or a pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight, or a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a dosage of pirfenidone or a pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 1200 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 800 mg to about 2400 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 1200 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 100 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 100 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 1200 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 100 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 800 mg to about 2400 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 100 ⁇ g of drug per -dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 1200 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 200 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 200 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 1200 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 200 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 800 mg to about 2400 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a v agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 200 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 1200 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a total weekly dosage of IFN-gamma containing an amount of about 100 ⁇ g to about 1,500 ⁇ g of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or administered substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a total weekly dosage of IFN-gamma containing an amount of about 100 ⁇ g to about 1,500 ⁇ g of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or administered substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 1200 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a total weekly dosage of IFN-gamma containing an amount of about 100 ⁇ g to about 1,500 ⁇ g of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or administered substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 800 mg to about 2400 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • the invention provides a method using an effective amount of IFN- ⁇ , pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a total weekly dosage of IFN-gamma containing an amount of about 100 ⁇ g to about 1,500 ⁇ g of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or administered substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 1200 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
  • a side effect management agent can be selected from the group of aNSAID (e.g., aspirin, ibuprofen, acetaminophen); a histamine type 2 receptor antagonist; a hematopoietic agent; and an antacid.
  • aNSAID e.g., aspirin, ibuprofen, acetaminophen
  • histamine type 2 receptor antagonist e.g., aspirin, ibuprofen, acetaminophen
  • a histamine type 2 receptor antagonist e.g., a histamine type 2 receptor antagonist
  • hematopoietic agent e.g., hematopoietic agent
  • an antacid e.g., an antacid.
  • Suitable dosages for the use of such side effect management agents are well known in the art.
  • the present invention provides combination therapy for the treatment of ALD, comprising administering an effective amount of a Type I interferon receptor agonist, an effective amount of a Type II interferon receptor agonist, and an effective amount of a TNF- ⁇ antagonist.
  • the method employs a Type II interferon receptor agonist that is IFN-gamma.
  • the method employs a Type I interferon receptor agonist that is IFN- ⁇ .
  • the methods further comprise administering an effective amount of pirfenidone or a pirfenidone analog.
  • the invention provides methods using a synergistically effective amount of a Type I interferon receptor agonist, a Type II interferon receptor agonist, and a TNF- ⁇ antagonist in the treatment of ALD in a patient.
  • the invention provides methods using a synergistically effective amount of an IFN- ⁇ , IFN- ⁇ , and a TNF- ⁇ antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRATM, in the treatment of ALD in a patient.
  • the invention provides a method using a synergistically effective amount of a consensus IFN- ⁇ , IFN- ⁇ , and a TNF- ⁇ antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRATM, in the treatment of ALD in a patient.
  • an effective amount of a consensus interferon (CIFN) and IFN- ⁇ suitable for use in the methods of the invention is provided by a dosage ratio of 1 ⁇ g CIFN : 10 ⁇ g IFN- ⁇ , where both CIFN and IFN- ⁇ are unPEGylated and unglycosylated species.
  • the invention provides a method using an effective amount of INFERGEN® consensus IFN- ⁇ , IF ⁇ - ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of INFERGEN® containing an amount of about 1 ⁇ g to about 30 ⁇ g, of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously bi
  • the invention provides a method using an effective amount of INFERGEN®consensus IFN- ⁇ , IF ⁇ - ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of INFERGEN® containing an amount of about 1 ⁇ g to about 9 ⁇ g, of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 100 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once montiily, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of
  • the invention provides a method using an effective amount of INFERGEN®consensus IFN- ⁇ , IFN- ⁇ and T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of INFERGEN® containing an amount of about 1 ⁇ g of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 50 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii)
  • the invention provides a method using an effective amount of INFERGEN®consensus IFN- ⁇ , IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient comprising administering to the patient a dosage of INFERGEN® containing an amount of about 9 ⁇ g of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN- ⁇ containing an amount of about 90 ⁇ g to about 100 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) RE
  • the invention provides a method using an effective amount of T ⁇ FERGEN®consensus IF ⁇ - ⁇ , IF ⁇ - ⁇ and T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of INFERGEN® containing an amount of about 30 ⁇ g of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN- ⁇ containing an amount of about 200 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw
  • the invention provides a method using an effective amount of PEGylated consensus IFN- ⁇ , IFN- ⁇ and T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEGylated consensus IFN- ⁇ (PEG-CIFN) containing an amount of about 4 ⁇ g to about 60 ⁇ g of CIFN amino acid weight per dose of PEG-CIFN, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN- ⁇ containing an amount of about 30 ⁇ g to about 1,000 ⁇ g of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/
  • the invention provides a method using an effective amount of PEGylated consensus IFN- ⁇ , IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEGylated consensus IFN- ⁇ (PEG-CIFN) containing an amount of about 18 ⁇ g to about 24 ⁇ g of CIFN amino acid weight per dose of PEG-CIFN, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN- ⁇ containing an amount of about 100 ⁇ g to about 300 ⁇ g of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg
  • an effective amount of IFN- ⁇ 2a or 2b or 2c and IFN- ⁇ suitable for use in the methods of the invention is provided by a dosage ratio of 1 million Units (MU) IFN- ⁇ 2a or 2b or 2c : 30 ⁇ g IFN- ⁇ , where both IFN- ⁇ 2a or 2b or 2c and IFN- ⁇ are unPEGylated and unglycosylated species.
  • MU 1 million Units
  • the invention provides a method using an effective amount of IFN- ⁇ 2a or 2b or 2c, IFN- ⁇ and T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IF ⁇ - ⁇ 2a, 2b or 2c containing an amount of about 1 MU to about 20 MU of drug per dose of IFN- ⁇ 2a, 2b or 2c subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, a dosage of IFN- ⁇ containing an amount of about 30 ⁇ g to about 600 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/
  • the invention provides a method using an effective amount of IFN- ⁇ 2a or 2b or 2c, IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN- ⁇ 2a, 2b or 2c containing an amount of about 3 MU of drug per dose of IFN- ⁇ 2a, 2b or 2c subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, a dosage of IFN- ⁇ containing an amount of about 100 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug infrav
  • the invention provides a method using an effective amount of IFN- ⁇ 2a or 2b or 2c, IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN- ⁇ 2a, 2b or 2c containing an amount of about 10 MU of drug per dose of IFN- ⁇ 2a, 2b or 2c subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, a dosage of IFN- ⁇ containing an amount of about 300 ⁇ g of drug per dose of IFN- ⁇ , subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously
  • the invention provides a method using an effective amount of PEGASYS®PEGylated IFN- ⁇ 2a, IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEGASYS® containing an amount of about 90 ⁇ g to about 360 ⁇ g, of drug per dose of PEGASYS®, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN- ⁇ containing an amount of about 30 ⁇ g to about 1,000 ⁇ g, of drug per week administered in divided doses subcutaneously qd, qod, tiw, or biw, or administered substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw,
  • the invention provides a method using an effective amount of PEGASYS®PEGylated IFN- ⁇ 2a, IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEGASYS® containing an amount of about 180 ⁇ g of drug per dose of PEGASYS®, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN- ⁇ containing an amount of about 100 ⁇ g to about 300 ⁇ g, of drug per week administered in divided doses subcutaneously qd, qod, tiw, or biw, or administered substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month
  • the invention provides a method using an effective amount of PEG-LNTRON®PEGylated IFN- ⁇ 2b, IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEG-INTRON® contaimng an amount of about 0.75 ⁇ g to about 3.0 ⁇ g of drug per kilogram of body weight per dose of PEG- INTRON®, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN- ⁇ containing an amount of about 30 ⁇ g to about 1,000 ⁇ g of drug per week administered in divided doses subcutaneously qd, qod, tiw, or biw, or administered substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about
  • the invention provides a method using an effective amount of PEG-INTRON®PEGylated IFN- ⁇ 2b, IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEG-INTRON® containing an amount of about 1.5 ⁇ g of drug per kilogram of body weight per dose of PEG-INTRON®, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN- ⁇ containing an amount of about 100 ⁇ g to about 300 ⁇ g of drug per week administered in divided doses subcutaneously qd, qod, tiw, or biw, or administered substantially continuously or continuously, and a dosage of a T ⁇ F- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously
  • the present invention provides for a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 9 ⁇ g INFERGEN® consensus IFN- ⁇ administered subcutaneously qd or tiw; 50 ⁇ g Actimmune® hximan IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow, for the desired treatment duration.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 9 ⁇ g INFERGEN® consensus IFN- ⁇ administered subcutaneously qd or tiw; 100 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/lcg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow, for the desired treatment duration.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 9 ⁇ g INFERGEN® consensus IFN- ⁇ administered subcutaneously qd or tiw; 25 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 9 ⁇ g INFERGEN® consensus IFN- ⁇ administered subcutaneously qd or tiw; 200 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 100 ⁇ g monoPEG(30 kD, linear)- ylated consensus IFN- ⁇ administered subcutaneously every 10 days or qw; 50 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to a patient a regimen of 100 ⁇ g monoPEG(30 kD, linear)- ylated consensus IFN- ⁇ administered subcutaneously every 10 days or qw; 100 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy. .
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 150 ⁇ g monoPEG(30 kD, linear)- ylated consensus IFN- ⁇ administered subcutaneously every 10 days or qw; 50 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 150 ⁇ g monoPEG(30 kD, linear)- ylated consensus IFN- ⁇ administered subcutaneously every 10 days or qw; 100 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 200 ⁇ g monoPEG(30 kD, linear)- ylated consensus IFN- ⁇ administered subcutaneously every 10 days or qw; 50 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 200 ⁇ g monoPEG(30 kD, linear)- ylated consensus IFN- ⁇ administered subcutaneously every 10 days or qw; 100 ⁇ g Actimmune® human IF ⁇ - ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered infravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy.
  • the therapeutic effect of the above regimens can be enhanced by co-administering to the patient a weight-based dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight, or a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg, or about 800 mg to about 2400 mg, or about 1000 mg to about 1800 mg, or about 1200 mg to about 1600 mg, orally qd for the desired duration of Type I interferon receptor agonist, Type II interferon receptor agonist and T ⁇ F- ⁇ antagonist therapy.
  • the present invention provides combination therapy for the treatment of ALD, comprising administering an effective amount of a Type II interferon receptor agonist, and an effective amount of a TNF- ⁇ antagonist.
  • the method employs a Type II interferon receptor agonist that is IF ⁇ -gamma.
  • the methods further comprise administering an effective amount of pirfenidone or a pirfenidone analog.
  • the invention provides methods using a synergistically effective amount of a Type II interferon receptor agonist and T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient.
  • the invention provides methods using a synergistically effective amount of IF ⁇ - ⁇ and a T ⁇ F- ⁇ antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRATM, in the treatment of ALD in a patient.
  • the invention provides a method using an effective amount of IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once
  • the invention provides a method using an effective amount of IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 100 ⁇ g of drug per dose of IFN- ⁇ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once
  • the invention provides a method using an effective amount of IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 10 ⁇ g to about 50 ⁇ g of drug per dose of IFN- ⁇ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once
  • the invention provides a method using an effective amount of IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 90 ⁇ g to about 100 ⁇ g of drug per dose of IFN- ⁇ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly
  • the invention provides a method using an effective amount of IFN- ⁇ and TNF- ⁇ antagonist in the freatment of ALD in a patient, comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 200 ⁇ g to about 300 ⁇ g of drug per dose of IFN- ⁇ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month
  • the invention provides a method using an effective amount of IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a total weekly dosage of IFN- ⁇ containing an amount of about 30 ⁇ g to about 1,000 ⁇ g of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or substantially continuously or continuously, and a dosage of a T ⁇ F- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the group consisting of (i) EN
  • the invention provides a method using an effective amount of IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a total weekly dosage of IFN- ⁇ containing an amount of about 100 ⁇ g to about 300 ⁇ g of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired
  • the invention provides a method using an effective amount of IFN- ⁇ and TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN- ⁇ containing an amount of about 30 ⁇ g to about 600 ⁇ g of drug per dose of IFN- ⁇ subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, and a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired
  • the therapeutic effect or other therapeutic benefit of such regimens can be enhanced by co-administering to the patient a weight-based dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight, or a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg, or about 800 mg to about 2400 mg, or about 1000 mg to about 1800 mg, or about 1200 mg to about 1600 mg, orally qd for the desired duration of Type II interferon receptor agonist and TNF- ⁇ antagonist therapy.
  • a Type II interferon receptor agonist and/or TNF- ⁇ antagonist is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time.
  • the duration of therapy with pirfenidone or a specific pirfenidone analog can be coincident with the duration of therapy with Type II interferon receptor agonist and/or TNF- ⁇ antagonist.
  • the present invention provides for a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 50 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow, for the desired treatment duration.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 100 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow, for the desired treatment duration.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 25 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of T ⁇ F- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy.
  • the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 200 ⁇ g Actimmune® human IFN- ⁇ lb administered subcutaneously tiw; and a dosage of TNF- ⁇ antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRATM subcutaneously qw or qow; for the desired duration of therapy.
  • the present invention provides combination therapy for the treatment of ALD, comprising administering an effective amount of a TNF- ⁇ antagonist and an effective amount of pirfenidone or a pirfenidone analog.
  • the invention provides methods using a synergistically effective amount of pirfenidone or a pirfenidone analog, and TNF- ⁇ antagonist in the treatment of ALD in a patient. In some embodiments, the invention provides methods using a synergistically effective amount of pirfenidone or a pirfenidone analog, and a T ⁇ F- ⁇ antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRATM, in the treatment of ALD in a patient.
  • the invention provides a method using a synergistically effective amount of pirfenidone, and a TNF- ⁇ antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRATM, in the treatment of ALD in a patient.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of a TNF- ⁇ antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration; and a dosage of pirfenidone or a pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the freatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired " treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drag subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising admimstering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising admimstering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a T ⁇ F- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drag intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfemdone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 1200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drag subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of aboutl200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 1200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1800 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drag subcutaneously biw, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1800 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drag intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF- ⁇ antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1800 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRATM in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
  • pirfenidone or a pirfenidone analog and/or a TNF- ⁇ antagonist is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time.
  • the duration of therapy with pirfenidone or a specific pirfenidone analog can be coincident with the duration of therapy with TNF- ⁇ antagonist.
  • the above-described methods of treatment of ALD are suitable for the treatment of any stage of ALD in a patient.
  • the subject methods can be employed in the treatment of hepatic steatosis, alcoholic hepatitis, hepatic fibrosis, or hepatic cirrhosis, or any combination thereof, that occurs in a patient suffering from ALD.
  • the above-described methods of treatment of ALD can be employed in the treatment of non-fibrotic alcoholic hepatitis in a patient.

Abstract

The present invention provides a method of treating alcoholic hepatitis, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in monotherapy or in combination therapy. The invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in combination therapy with a second therapeutic agent other than IFN-Ϝ. The invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog, a Type II interferon receptor agonist, and a side effect management agent. The invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of a TNF-α antagonist, which therapy further includes administration of an effective amount of a Type II interferon receptor agonist, and/or pirfenidone or a pirfenidone analog.

Description

METHOD OF TREATING ALCOHOLIC LIVER DISEASE
FIELD OF THE INVENTION [0001] The present invention is in the field of alcoholic liver disease, and use of pirfenidone or a pirfenidone analog to treat alcoholic liver disease.
BACKGROUND OF THE INVENTION
[0002] Alcoholic liver disease (ALD) is a major cause of illness and death, and is the most common liver disease in the United States. It is the fourth leading cause of death in the United States, and results in between 20,000 and 40,000 deaths per year. Women are generally more susceptible to alcohol-induced liver damage than men and develop alcoholic liver disease at a more rapid rate having imbibed less alcohol.
[0003] ALD involves an acute or chronic inflammation of the liver induced by alcohol abuse. ALD is characterized by fatty liver (steatosis), hepatitis, liver fibrosis, and cirrhosis. Alcoholic hepatitis is characterized histologically by hepatocellular necrosis, alcoholic Mallory's hyaline bodies, and an inflammatory reaction with infiltration by polymorphonuclear leukocytes and lymphocytes. The clinical presentation of alcoholic hepatitis varies with the severity of the disease. Common symptoms are weakness, anorexia, weight loss, nausea, vomiting, and diarrhea. Patients often present with fever, jaundice, and tender hepatomegaly.
[0004] There is a need in the art for methods of treating ALD. The present invention addresses this need. Literature
[0005] U.S. Patent No. 5,952,309; and Woods et al. (1993) Am. Fam. Physician Apr;47(5): 1171-8.
SUMMARY OF THE INVENTION [0006] The present invention provides a method of treating alcoholic hepatitis, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in monofherapy or in combination therapy. The invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in combination therapy with a second therapeutic agent other than IFN-γ. The invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog, a Type II interferon receptor agonist, and a side effect management agent (e.g., a palliative agent, or an agent for the treatment, reduction, or avoidance of a side effect caused by any therapeutic agent). The invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of a TNF- antagonist, which therapy further includes administration of an effective amount of a Type II interferon receptor agonist, and/or pirfenidone or a pirfenidone analog.
DEFINITIONS
[0007] The terms "individual," "host," "subject," and "patient" are used interchangeably herein, and refer to a mammal, including, but not limited to, primates, including simians and humans.
[0008] As used herein, the terms "treatment," "treating," and the like, refer to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse affect attributable to the disease. "Treatment," as used herein, covers any treatment of a disease in a mammal, particularly in a human, and includes: (a) preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; and (c) relieving the disease, i.e., causing regression of the disease.
[0009] As used herein, the term "alcoholic hepatitis," refers to an acute or chronic inflammatory lesion of the liver that occurs in the context of chronic alcohol abuse.
[0010] As used herein, the term "alcoholic hepatic fibrosis," used interchangeably herein with "alcoholic liver fibrosis," refers to the growth of scar tissue in the liver that can occur in the context of chronic alcohol abuse.
[0011] As used herein, the term "liver function" refers to a normal function of the liver, including, but not limited to, a synthetic function, including, but not limited to, synthesis of proteins such as serum proteins (e.g., albumin, clotting factors, alkaline phosphatase, aminotransferases (e.g., alanine transaminase, aspartate transaminase), 5'-nucleosidase, γ- glutaminyltranspeptidase, etc.), synthesis of bilirubin, synthesis of cholesterol, and synthesis of bile acids; a liver metabolic function, including, but not limited to, carbohydrate metabolism, amino acid and ammonia metabolism, hormone metabolism, and lipid metabolism; detoxification of exogenous drugs; a hemodynamic function, including splanchnic and portal hemodynamics; and the like. [0012] The term "dosing event" as used herein refers to administration of a therapeutic agent to a patient in need thereof, which event may encompass one or more releases of agent from a drug dispensing device.
[0013] "Continuous delivery" as used herein (e.g. , in the context of "continuous delivery of a substance to a tissue") is meant to refer to movement of drug to a delivery site, e.g., into a tissue in a fashion that provides for delivery of a desired amount of substance into the tissue over a selected period of time, where about the same quantity of drug is received by the patient each minute during the selected period of time.
[0014] "Controlled release" as used herein (e.g., in the context of "controlled drug release") is meant to encompass release of substance (e.g., a Type II interferon receptor agonist, e.g., IFN- γ) at a selected or otherwise controllable rate, interval, and/or amount, which is not substantially influenced by the environment of use. "Controlled release" thus encompasses, but is not necessarily limited to, substantially continuous delivery, and patterned delivery (e.g., intermittent delivery over a period of time that is interrupted by regular or irregular time intervals).
[0015] "Patterned" or "temporal" as used in the context of drug delivery means delivery of drug in a pattern, generally a substantially regular pattern, over a pre-selected period of time (e.g. , other than a period associated with, for example a bolus injection). "Patterned" or "temporal" drug delivery is meant to encompass delivery of drug at an increasing, decreasing, substantially constant, or pulsatile, rate or range of rates (e.g., amount of drug per unit time, or volume of drug formulation for a unit time), and further encompasses delivery that is continuous or substantially continuous, or chronic.
[0016] The term "controlled drug delivery device" is meant to encompass any device wherein the release (e.g., rate, timing of release) of a drug or other desired substance contained therein is controlled by or determined by the device itself and not substantially influenced by the environment of use, or releasing at a rate that is reproducible within the environment of use.
[0017] By "substantially continuous" as used in, for example, the context of "substantially continuous infusion" or "substantially continuous delivery" is meant to refer to delivery of drug in a manner that is substantially uninterrupted for a pre-selected period of drug delivery, where the quantity of drug received by the patient during any 8 hour interval in the pre-selected period never falls to zero. Furthermore, "substantially continuous" drug delivery can also encompass delivery of drug at a substantially constant, pre-selected rate or range of rates (e.g., amount of drug per unit time, or volume of drug formulation for a unit time) that is substantially uninterrupted for a pre-selected period of drug delivery. [0018] As used herein, the term "pirfenidone" means 5-methyl-l-phenyl-2-(lH)-pyridone. As used herein, the term "pirfenidone analog" means any compound of Formula I, IIA or IIB below. As used herein, the term "specific pirfenidone analog" refers to, and is limited to, each and every pirfenidone analog shown in Table 1.
[0019] As used herein, the term "a Type I interferon receptor agonist" refers to any naturally occurring or non-naturally occurring ligand of human Type I interferon receptor, which binds to and causes signal transduction via the receptor. Type I interferon receptor agonists include interferons, including naturally-occurring interferons, modified mterferons, synthetic interferons, pegylated interferons, fusion proteins comprising an interferon and a heterologous protein, shuffled interferons; antibody specific for an interferon receptor; non-peptide chemical agonists; and the like.
[0020] As used herein, the tenn "a Type II interferon receptor agonist" refers to any naturally- occurring or non-naturally-occurring ligand of a human Type II interferon receptor which binds to and causes signal transduction via the receptor. Type II interferon receptor agonists include interferons, including naturally-occurring interferons, modified interferons, synthetic interferons, pegylated interferons, fusion proteins comprising an interferon and a heterologous protein, shuffled interferons; antibody specific for an interferon receptor; non-peptide chemical agonists; and the like.
[0021] As used herein, any compound or agent described as "effective for the avoidance or amelioration of side effects induced by a Type II interferon receptor agonist and/or pirfenidone or a pirfenidone analog," or as "effective for reducing or eliminating the severity or occurrence of side effects induced by a Type II interferon receptor agonist and/or pirfenidone or a pirfenidone analog," or any compound or agent described by language with a meaning similar or equivalent to that of either of the foregoing quoted passages, is/are defined as a compound(s) or agent(s) that when co-administered to a patient in an effective amount along with a given dosing regimen of a Type II interferon receptor agonist and/or pirfenidone or a pirfenidone analog, abates or eliminates the severity or occurrence of side effects experienced by a patient in response to the given dosing regimen of the Type II interferon receptor agonist and/or pirfenidone or pirfenidone analog therapy, as compared to the severity or occurrence of side effects that would have been experienced by the patient in response to the same dosing regimen of the Type II interferon receptor agonist and/or pirfenidone or pirfenidone analog therapy without co-administration of the agent.
[0022] In many embodiments, the effective amounts of a Type II interferon receptor agonist and pirfenidone or a pirfenidone analog are synergistic amounts. As used herein, a "synergistic combination" or a "synergistic amount" of a Type II interferon re'ceptor agonist and pirfenidone or a pirfenidone analog is a combination or amount that is more effective in the therapeutic or prophylactic treatment of ALD and/or alcoholic hepatitis than the incremental improvement in treatment outcome that could be predicted or expected from a merely additive combination of (i) the therapeutic or prophylactic benefit of the Type II interferon receptor agonist when administered at that same dosage as a monotherapy and (ii) the therapeutic or prophylactic benefit of the pirfenidone or a pirfenidone analog when administered at the same dosage as a monotherapy.
[0023] Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
[0024] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.
[0025] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.
[0026] It must be noted that as used herein and in the appended claims, the singular forms "a," "and," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "a therapeutic agent" includes a plurality of such agents and reference to "the liver function" includes reference to one or more liver functions and equivalents thereof known to those skilled in the art, and so forth. [0027] The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed.
DETAILED DESCRIPTION OF THE INVENTION [0028] The present invention provides a method of treating alcoholic hepatitis, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in monotherapy or in combination therapy. The invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in combination therapy with a second therapeutic agent other than IFN-γ. The invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog, a Type II interferon receptor agonist, and a side effect management agent (e.g., a palliative agent, or an agent for the treatment, reduction, or avoidance of a side effect caused by any therapeutic agent). The invention further provides a method of treating alcoholic liver disease, the method involving administering to an individual in need thereof an effective amount of a TNF-α antagonist, which therapy further includes administration of an effective amount of a Type II interferon receptor agonist, and/or pirfenidone or a pirfenidone analog. TREATMENT METHODS [0029] As used herein, the term "alcoholic liver disease" or "ALD" includes hepatic steatosis, alcoholic hepatitis, hepatic fibrosis, and hepatic cirrhosis, which occur as the result of chronic alcohol abuse. The present invention provides methods of treating ALD, involving administering one or more therapeutic agents, where the monotherapy or combination therapy is effective to ameliorate one or more of hepatic steatosis, alcoholic hepatitis, hepatic fibrosis, and hepatic cirrhosis. The present invention provides method of treating alcoholic hepatitis, involving administering one or more therapeutic agents, where the monotherapy or combination therapy is effective to ameliorate alcoholic hepatitis. Non-Fibrotic Alcoholic Hepatitis [0030] The present invention provides a method of treating early stage ALD, e.g., treating alcoholic hepatitis in the absence of hepatic fibrosis ("non-fibrotic alcoholic hepatitis"), the method involving administering to an individual in need thereof an effective amount of pirfenidone or a pirfenidone analog in monotherapy or in combination therapy.
[0031] In some embodiments, an "effective amount" of pirfenidone or a pirfenidone analog is an amount or a dosage that, when administered to an individual in monotherapy, is effective to treat non-fibrotic alcoholic hepatitis, e.g., is effective to reduce at least one symptom and/or histological characteristic of alcoholic hepatitis.
[0032] In some embodiments, an effective amount of pirfenidone or a pirfenidone analog is an amount that is effective to cause a reduction in one or more symptoms of non-fibrotic alcoholic hepatitis of at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90% or more, compared with the level or degree of the symptom in the absence of treatment with pirfenidone or pirfenidone analog. Symptoms of non-fibrotic alcoholic hepatitis include fever, jaundice, and tender hepatomegaly.
[0033] In some embodiments, an effective amount of pirfenidone or a pirfenidone analog is an amount that is effective to cause a reduction in one or more histological characteristics of non- fibrotic alcoholic hepatitis of at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or at least about 90% or more, compared with the histological characteristic in the absence of treatment with pirfenidone or a pirfenidone analog. Histological characteristics of non-fibrotic alcoholic hepatitis include hepatocellular necrosis, alcoholic Mallory's hyaline bodies, and inflammatory lesions with infiltrating polymorphonuclear leukocytes and lymphocytes.
[0034] In some embodiments, an effective amount of pirfenidone or a pirfenidone analog is an amount that is effective to increase an index of liver function by at least about 10%, at least about 20%, at least about 25%, at least about 30%), at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%, or more, compared to the index of liver function in an untreated individual, or in a placebo-treated individual. Those skilled in the art can readily measure such indices of liver function, using standard assay methods, many of which are commercially available, and are used routinely in clinical settings. 0035] In some embodiments, a subject method for treating non-fibrotic alcoholic hepatitis comprises administering an effective amount of pirfenidone or a pirfenidone analog, and an effective amount of at least a second therapeutic agent. Suitable second therapeutic agents include, but are not limited to a Type II interferon receptor agonist, and a TNF antagonist. "At least a second therapeutic agent" includes: a Type II interferon receptor agonist; a TNF antagonist; a Type I interferon receptor agonist and a Type II interferon receptor agonist; a Type II interferon receptor agonist and a TNF antagonist; and a Type II interferon receptor agonist, a Type I interferon receptor agonist and a TNF antagonist.
[0036] In some embodiments, "effective amounts" of pirfenidone or a pirfenidone analog and at least a second therapeutic agent are combined amounts that, when administered to an individual in combination therapy, are effective to treat non-fibrotic alcoholic hepatitis, e.g., is effective to reduce at least one symptom and/or histological characteristic of alcoholic hepatitis.
[0037] In some embodiments, "effective amounts" of pirfenidone or a pirfenidone analog and at least a second therapeutic agent are combined amounts that, when administered to an individual in combination therapy, are effective to cause a reduction in one or more symptoms of non-fibrotic alcoholic hepatitis of at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%), or at least about 90% or more, compared with the level or degree of the symptom in the absence of treatment with the combination therapy. Symptoms of non-fibrotic alcoholic hepatitis include fever, jaundice, and tender hepatomegaly.
[0038] In some embodiments, "effective amounts" of pirfenidone or a pirfenidone analog and at least a second therapeutic agent are combined amounts that, when administered to an individual in combination therapy, are effective to cause a reduction in one or more histological characteristics of non-fibrotic alcoholic hepatitis of at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%>, at least about 70%, at least about 80%, or at least about 90% or more, compared with the histological characteristic in the absence of treatment with the combination therapy. Histological characteristics of non-fibrotic alcoholic hepatitis include hepatocellular necrosis, alcoholic Mallory's hyaline bodies, and inflammatory lesions with infiltrating polymorphonuclear leukocytes and lymphocytes. )039] In some embodiments, "effective amounts" of pirfenidone or a pirfenidone analog and at least a second therapeutic agent are combined amounts that, when administered to an individual in combination therapy, are effective to increase an index of liver function by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 10%, at least about 75%, or at least about 80%, or more, compared to the index of liver function in an untreated individual, or in a placebo-treated individual. Those skilled in the art can readily measure such indices of liver function, using standard assay methods, many of which are commercially available, and are used routinely in clinical settings. Alcoholic liver disease
[0040] The present invention provides methods of treating alcoholic liver disease, including reducing alcoholic hepatitis, reducing clinical liver fibrosis, reducing the likelihood that liver fibrosis will occur, reducing a parameter associated with liver fibrosis, and reducing liver cirrhosis.
[0041] In some embodiments, the methods involve administering an effective amount of pirfenidone or a pirfenidone analog in monotherapy. In some embodiments, the methods involve administering an effective amount of pirfenidone or a pirfenidone analog in combination therapy with an effective amount of a second therapeutic agent other than IFN-γ (e.g., where the second therapeutic agent is a TNF antagonist). In some embodiments, the methods involve administering an effective amount of pirfenidone or a pirfenidone analog, an effective amount of a Type II interferon receptor agonist, and a side effect management agent that reduces at least one side effect of a pirfenidone or a pirfenidone analog and/or a Type II interferon receptor agonist. In some embodiments, the methods involve co-administering an effective amount a TNF antagonist and an effective amount of a Type II interferon receptor agonist; in some of these embodiments, the methods further involve administering pirfenidone or a pirfenidone analog. In some embodiments, the methods involve administering an effective amount of a TNF antagonist, an effective amount of a Type II interferon receptor agonist, and an effective amount of a Type I interferon receptor agonist; in some of these embodiments, the methods further involve administering pirfenidone or a pirfenidone analog. In some embodiments, the methods involve administering an effective amount of a TNF antagonist and an effective amount of pirfenidone or a pirfenidone analog. Of particular interest in many embodiments is treatment of humans.
[0042] Alcoholic liver fibrosis is a precursor to liver cirrhosis. Accordingly, the present invention further provides methods of reducing the likelihood that an individual will develop liver cirrhosis. Alcoholic liver fibrosis is a precursor to the complications associated with liver cirrhosis, such as portal hypertension, progressive liver insufficiency, and hepatocellular carcinoma. A reduction in liver fibrosis thus reduces the incidence of such complications. Accordingly, the present invention further provides methods of reducing the likelihood that an individual will develop complications associated with cirrhosis of the liver.
[0043] As used herein, "effective amounts" of a therapeutic agent (e.g., a Type II interferon receptor agonist, a Type I interferon receptor agonist, a TNF-α antagonist, pirfenidone or a pirfenidone analog, or any of the above-mentioned combinations thereof) are any combined dosage that is effective in reducing liver fibrosis or reduce the rate of progression of alcoholic liver fibrosis; and/or that is effective in reducing the likelihood that an individual will develop alcoholic liver fibrosis; and/or that is effective in reducing a parameter associated with alcoholic liver fibrosis; and/or that is effective in reducing a disorder associated with ciπhosis of the liver.
[0044] The invention also provides a method for treatment of alcoholic liver fibrosis in an individual comprising one or more therapeutic agents as described above in monotherapy or in combination therapy in amounts that are effective for prophylaxis or therapy of liver fibrosis in the individual, e.g., increasing the probability of survival, reducing the risk of death, ameliorating the disease burden or slowing the progression of disease in the individual.
[0045] Whether a subject monotherapy or combination therapy is effective in reducing alcoholic liver fibrosis can be determined by any of a number of well-established techniques for measuring liver fibrosis and liver function. Whether liver fibrosis is reduced is determined by analyzing a liver biopsy sample. An analysis of a liver biopsy comprises assessments of two major components: necroinflammation assessed by "grade" as a measure of the severity and ongoing disease activity, and the lesions of fibrosis and parenchymal or vascular remodeling as assessed by "stage" as being reflective of long-term disease progression. See, e.g., Brunt (2000) Hepatol. 31:241-246; and METAVIR (1994) Hepatology 20:15-20. Based on analysis of the liver biopsy, a score is assigned. A number of standardized scoring systems exist which provide a quantitative assessment of the degree and severity of fibrosis. These include the METAVIR, Knodell, Scheuer, Ludwig, and Ishak scoring systems.
[0046] The METAVIR scoring system is based on an analysis of various features of a liver biopsy, including fibrosis (portal fibrosis, centrilobular fibrosis, and cirrhosis); necrosis (piecemeal and lobular necrosis, acidophilic retraction, and ballooning degeneration); inflammation (portal tract inflammation, portal lymphoid aggregates, and distribution of portal inflammation); bile duct changes; and the Knodell index (scores of periportal necrosis, lobular necrosis, portal inflammation, fibrosis, and overall disease activity). The definitions of each stage in the METAVIR system are as follows: score: 0, no fibrosis; score: 1, stellate enlargement of portal tract but without septa formation; score: 2, enlargement of portal tract with rare septa formation; score: 3, numerous septa without cirrhosis; and score: 4, cirrhosis.
[0047] Knodell's scoring system, also called the Hepatitis Activity Index, classifies specimens based on scores in four categories of histologic features: I. Periportal and/or bridging necrosis; II. Intralobular degeneration and focal necrosis; III. Portal inflammation ; and IV. Fibrosis. In the Knodell staging system, scores are as follows: score: 0, no fibrosis; score: 1, mild fibrosis (fibrous portal expansion); score: 2, moderate fibrosis; score: 3, severe fibrosis (bridging fibrosis); and score: 4, cirrhosis. The higher the score, the more severe the liver tissue damage. Knodell (1981) Hepatol. 1:431.
[0048] In the Scheuer scoring system scores are as follows: score: 0, no fibrosis; score: 1, enlarged, fibrotic portal tracts; score: 2, periportal or portal-portal septa, but intact architecture; score: 3, fibrosis with architectural distortion, but no obvious cirrhosis; score: 4, probable or definite cirrhosis. Scheuer (1991) J Hepatol. 13:372.
[0049] The Ishak scoring system is described in Ishak (1995) J Hepatol. 22:696-699. Stage 0, No fibrosis; Stage 1, Fibrous expansion of some portal areas, with or without short fibrous septa; stage 2, Fibrous expansion of most portal areas, with or without short fibrous septa; stage 3, Fibrous expansion of most portal areas with occasional portal to portal (P-P) bridging; stage 4, Fibrous expansion of portal areas with marked bridging (P-P) as well as portal-central (P-C); stage 5, Marked bridging (P-P and/or P-C) with occasional nodules (incomplete cirrhosis); stage 6, Cirrhosis, probable or definite .
[0050] The benefit of anti-fibrotic therapy can also be measured and assessed by using the Child-Pugh scoring system which comprises a multicomponent point system based upon abnormalities in serum bilirubin level, serum albumin level, prothrombin time, the presence and severity of ascites, and the presence and severity of encephalopathy. Based upon the presence and severity of abnormality of these parameters, patients may be placed in one of three categories of increasing severity of clinical disease: A, B, or C.
[0051] In some embodiments, therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that effects a change of one unit or more in the fibrosis stage based on pre- and post- therapy liver biopsies. In particular embodiments, therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) reduce liver fibrosis by at least one unit in the METAVIR, the Knodell, the Scheuer, the Ludwig, or the Ishak scoring system.
[0052] Secondary, or indirect, indices of liver function can also be used to evaluate the efficacy of treatment with the subject therapy. Morphometric computerized semi-automated assessment of the quantitative degree of liver fibrosis based upon specific staining of collagen and/or serum markers of liver fibrosis can also be measured as an indication of the efficacy of a subject treatment method. Secondary indices of liver function include, but are not limited to, serum transaminase levels, prothrombin time, bilirubin, platelet count, portal pressure, albumin level, and assessment of the Child-Pugh score.
[0053] In another embodiment, therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that are effective to increase an index of liver function by at least about 10%, at least about 20%), at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%>, at least about 65%, at least about 70%, at least about 75%, or at least about 80%>, or more, compared to the index of liver function in an untreated individual, or in a placebo-treated individual. Those skilled in the art can readily measure such indices of liver function, using standard assay methods, many of which are commercially available, and are used routinely in clinical settings.
[0054] Serum markers of liver fibrosis can also be measured as an indication of the efficacy of a subject treatment method. Serum markers of liver fibrosis include, but are not limited to, hyaluronate, N-terminal procollagen III peptide, 7S domain of type IV collagen, C-terminal procollagen I peptide, and laminin. Additional biochemical markers of liver fibrosis include α- 2-macroglobulin, haptoglobin, gamma globulin, apolipoprotein A, and gamma glutamyl transpeptidase.
[0055] In another embodiment, therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that are effective to reduce a serum level of a marker of liver fibrosis by at least about 10%), at least about 20%, at least about 25%>, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%>, at least about 55%>, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%, or more, compared to the level of the marker in an untreated individual, or in a placebo-treated individual. Those skilled in the art can readily measure such serum markers of liver fibrosis, using standard assay methods, many of which are commercially available, and are used routinely in clinical settings. Methods of measuring serum markers include immunological- based methods, e.g., enzyme-linked immunosorbent assays (ELISA), radioimmunoassays, and the like, using antibody specific for a given serum marker.
[0056] Quantitative tests of functional liver reserve can also be used to assess the efficacy of treatment with the subject therapy. These include: indocyanine green clearance (ICG), galactose elimination capacity (GEC), aminopyrine breath test (ABT), antipyrine clearance, monoethylglycine-xylidide (MEG-X) clearance, and caffeine clearance. [0057] As used herein, a "complication associated with cirrhosis of the liver" refers to a disorder that is a sequellae of decompensated liver disease, i.e., or occurs subsequently to and as a result of development of liver fibrosis, and includes, but is not limited to, development of ascites, variceal bleeding, portal hypertension, jaundice, progressive liver insufficiency, encephalopathy, hepatocellular carcinoma, liver failure requiring liver transplantation, and liver-related mortality.
[0058] In another embodiment, therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that is effective in reducing the incidence of (e.g., the likelihood that an individual will develop) a disorder associated with cirrhosis of the liver by at least about 10%, at least about 20%, at least about 25%, at least about 30%>, at least about 35%, at least about 40%, at least about 45%>, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, or at least about 80%, or more, compared to an untreated individual, or in a placebo-treated individual.
[0059] Whether a subject monotherapy or combination therapy is effective in reducing the incidence of a disorder associated with cirrhosis of the liver can readily be determined by those skilled in the art.
[0060] Reduction in liver fibrosis increases liver function. Thus, in another embodiment, therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that is effective in increasing liver function. Liver functions include, but are not limited to, synthesis of proteins such as serum proteins (e.g., albumin, clotting factors, alkaline phosphatase, aminotransferases (e.g., alanine transaminase, aspartate transaminase), 5'-nucleosidase, γ- glutaminyltranspeptidase, etc.), synthesis of bilirubin, synthesis of cholesterol, and synthesis of bile acids; a liver metabolic function, including, but not limited to, carbohydrate metabolism, amino acid and ammonia metabolism, hormone metabolism, and lipid metabolism; detoxification of exogenous drugs; a hemodynamic function, including splanchnic and portal hemodynamics; and the like.
[0061] Whether a liver function is increased is readily ascertainable by those skilled in the art, using well-established tests of liver function. Thus, synthesis of markers of liver function such as albumin, alkaline phosphatase, alanine transaminase, aspartate transaminase, bilirubin, and the like, can be assessed by measuring the level of these markers in the serum, using standard immunological and enzymatic assays. Splanchnic circulation and portal hemodynamics can be measured by portal wedge pressure and/or resistance using standard methods. Metabolic functions can be measured by measuring the level of ammonia in the serum.
[0062] Whether serum proteins normally secreted by the liver are in the normal range can be determined by measuring the levels of such proteins, using standard immunological and enzymatic assays. Those skilled in the art know the normal ranges for such serum proteins. The following are non-limiting examples. The normal range of alanine transaminase is from about 7 to about 56 units per liter of serum. The normal range of aspartate transaminase is from about 5 to about 40 units per liter of serum. Bilirubin is measured using standard assays. Normal bilirubin levels are usually less than about 1.2 mg/dL. Serum albumin levels are measured using standard assays. Normal levels of serum albumin are in the range of from about 35 to about 55 g/L. Prolongation of prothrombin time is measured using standard assays. Normal prothrombin time is less than about 4 seconds longer than control.
[0063] In another embodiment, therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that is effective to increase liver function by at least about 10%), at least about 20%), at least about 30%, at least about 40%, at least about 50%>, at least about 60%, at least about 70%, at least about 80%, or more, compared to an untreated individual, or in a placebo-treated individual. For example, in some embodiments, therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) that is effective to reduce an elevated level of a serum marker of liver function by at least about 10%>, at least about 20%, at least about 30%, at least about 40%, at least about 50%), at least about 60%>, at least about 70%, at least about 80%, or more, or to reduce the level of the serum marker of liver function to within a normal range. In other embodiments, therapeutically effective amounts of a therapeutic agent are any dosage (e.g., a dosage in a subject monotherapy or a combined dosage in a subject combination therapy) effective to increase a reduced level of a serum marker of liver function by at least about 10%, at least about 20%), at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%), at least about 80%, or more, or to increase the level of the serum marker of liver function to within a normal range. Type I interferon receptor agonists
[0064] In some of the above-described methods, a Type I interferon receptor agonist is administered. Type I interferon receptor agonists include an IFN-α; an IFN-β; an IFN-tau; an IFN-ω; antibody agonists specific for a Type I interferon receptor; and any other agonist of Type I interferon receptor, including non-polypeptide agonists. Interferon-Alpha
[0065] Any known IFN-α can be used in the instant invention. The term "interferon-alpha" as used herein refers to a family of related polypeptides that inhibit viral replication and cellular proliferation and modulate immune response. The term "IFN-α" includes naturally occurring IFN-α; synthetic IFN-α; derivatized IFΝ-α (e.g., PEGylated IFN-α, glycosylated IFN-α, and the like); and analogs of naturally occurring or synthetic IFN-α; essentially any IFN-α that has antiviral properties, as described for naturally occurring IFN-α.
[0066] Suitable alpha interferons include, but are not limited to, naturally-occurring IFN-α (including, but not limited to, naturally occurring IFN-α2a, IFN-α2b); recombinant interferon alpha-2b such as Intron-A interferon available from Schering Corporation, Kenilworth, N.J.; recombinant interferon alpha-2a such as Roferon interferon available from Hoffmann-La Roche, Nutley, N. J.; recombinant interferon alpha-2C such as Berofor alpha 2 interferon available from Boehringer Ingelheim Pharmaceutical, Inc., Ridgefield, Conn.; interferon alpha- nl, a purified blend of natural alpha interferons such as Sumiferon available from Sumitomo, Japan or as Wellferon interferon alpha-nl (INS) available from the Glaxo-Wellcome Ltd., London, Great Britain; and interferon alpha-n3 a mixture of natural alpha interferons made by Interferon Sciences and available from the Purdue Frederick Co., Norwalk, Conn., under the Alferon Tradename.
[0067] The term "IFN-α" also encompasses consensus IFN-α. Consensus IFΝ-α (also referred to as "CIFN" and "IFN-con" and "consensus interferon") encompasses but is not limited to the amino acid sequences designated IFN-coni, IFN-con2 and IFN-con3 which are disclosed in U.S. Pat. Nos. 4,695,623 and 4,897,471; and consensus interferon as defined by determination of a consensus sequence of naturally occurring interferon alphas (e.g., Infergen®, InterMune, Inc., Brisbane, Calif). IFN-coni is the consensus interferon agent in the Infergen® alfacon-1 product. The Infergen® consensus interferon product is referred to herein by its brand name (Infergen®) or by its generic name (interferon alfacon-1). DNA sequences encoding IFN-con may be synthesized as described in the aforementioned patents or other standard methods. Use of CIFN is of particular interest.
[0068] Also suitable for use in the present invention are fusion polypeptides comprising an IFN-α and a heterologous polypeptide. Suitable IFN-α fusion polypeptides include, but are not limited to, Albuferon-alpha™ (a fiision product of human albumin and IFN-α; Human Genome Sciences; see, e.g., Osborn et al. (2002) J Pharmacol. Exp. Therap. 303:540-548). Also suitable for use in the present invention are gene-shuffled forms of IFN-α. See., e.g., Masci et al. (2003) Curr. Oncol. Rep. 5:108-113. PEGylated Interferon-Alpha
[0069] The term "IFN-α" also encompasses derivatives of IFN-α that are derivatized (e.g., are chemically modified) to alter certain properties such as serum half-life. As such, the term "IFN-α" includes glycosylated IFN-α; IFΝ-α derivatized with polyethylene glycol ("PEGylated IFN-α"); and the like. PEGylated IFN-α, and methods for making same, is discussed in, e.g., U.S. Patent Nos. 5,382,657; 5,981,709; and 5,951,974. PEGylated IFN-α encompasses conjugates of PEG and any of the above-described IFN-α molecules, including, but not limited to, PEG conjugated to interferon alpha-2a (Roferon, Hoffman La-Roche, Nutley, N. J.), interferon alpha 2b (Intron, Schering-Plough, Madison, N. J.), interferon alpha-2c (Berofor Alpha, Boehringer Ingelheim, Ingelheim, Germany); and consensus interferon as defined by determination of a consensus sequence of naturally occurring interferon alphas (Infergen®, InterMune, Inc., Brisbane, Calif).
[0070] Any of the above-mentioned IFN-α polypeptides can be modified with one or more polyethylene glycol moieties, i.e., PEGylated. The PEG molecule of a PEGylated IFN-α polypeptide is conjugated to one or more amino acid side chains of the IFN-α polypeptide. In some embodiments, the PEGylated IFN-α contains a PEG moiety on only one amino acid. In other embodiments, the PEGylated IFN-α contains a PEG moiety on two or more amino acids, e.g., the IFN-α contains a PEG moiety attached to two, three, four, five, six, seven, eight, nine, or ten different amino acid residues.
[0071] IFN-α may be coupled directly to PEG (i.e., without a linking group) through an amino group, a sulfhydryl group, a hydroxyl group, or a carboxyl group.
[0072] In some embodiments, the PEGylated IFN-α is PEGylated at or near the amino terminus (Ν-terminus) of the IFN-α polypeptide, e.g., the PEG moiety is conjugated to the IFN-α polypeptide at one or more amino acid residues from amino acid 1 through amino acid 4, or from amino acid 5 through about 10.
[0073] In other embodiments, the PEGylated IFN-α is PEGylated at one or more amino acid residues from about 10 to about 28.
[0074] In other embodiments, the PEGylated IFN-α is PEGylated at or near the carboxyl terminus (C-terminus) of the IFN-α polypeptide, e.g., at one or more residues from amino acids 156-166, or from amino acids 150 to 155.
[0075] In other embodiments, the PEGylated IFN-α is PEGylated at one or more amino acid residues at one or more residues from amino acids 100-114.
[0076] The polyethylene glycol derivatization of amino acid residues at or near the receptor- binding and/or active site domains of the IFN-α protein can disrupt the functioning of these domains. In certain embodiments of the invention, amino acids at which PEGylation is to be avoided include amino acid residues from amino acid 30 to amino acid 40; and amino acid residues from amino acid 113 to amino acid 149.
[0077] In some embodiments, PEG is attached to IFN-α via a linking group. The linking group is any biocompatible linking group, where "biocompatible" indicates that the compound or group is non-toxic and may be utilized in vitro or in vivo without causing injury, sickness, disease, or death. PEG can be bonded to the linking group, for example, via an ether bond, an ester bond, a thiol bond or an amide bond. Suitable biocompatible linking groups include, but are not limited to, an ester group, an amide group, an imide group, a carbamate group, a carboxyl group, a hydroxyl group, a carbohydrate, a succinimide group (including, for example, succinimidyl succinate (SS), succinimidyl propionate (SPA), succinimidyl butanoate (SBA), succinimidyl carboxymethylate (SCM), succinimidyl succinamide (SSA) or N-hydroxy succinimide (NHS)), an epoxide group, an oxycarbonylimidazole group (including, for example, carbonyldimidazole (GDI)), a nitro phenyl group (including, for example, nitrophenyl carbonate (ΝPC) or trichlorophenyl carbonate (TPC)), a trysylate group, an aldehyde group, an isocyanate group, a vinylsulfone group, a tyrosine group, a cysteine group, a histidine group or a primary amine.
[0078] Methods for making succinimidyl propionate (SPA) and succinimidyl butanoate (SBA) ester-activated PEGs are described in U.S. Pat. No. 5,672,662 (Harris, et al.) and WO 97/03106.
[0079] Methods for attaching a PEG to an IFΝ-α polypeptide are known in the art, and any known method can be used. See, for example, by Park et al, Anticancer Res., 1 :373-376 (1981); Zaplipsky and Lee, Polyethylene Glycol Chemistry: Biotechnical and Biomedical Applications, J. M. Harris, ed., Plenum Press, NY, Chapter 21 (1992); U.S. Patent No. 5,985,265; U.S. Pat. No. 5,672,662 (Harris, et al.) and WO 97/03106.
[0080] Pegylated IFN-α, and methods for making same, is discussed in, e.g., U.S. Patent Nos. 5,382,657; 5,981,709; 5,985,265; and 5,951,974. Pegylated IFN-α encompasses conjugates of PEG and any of the above-described IFN-α molecules, including, but not limited to, PEG conjugated to interferon alpha-2a (Roferon, Hoffman LaRoche, Nutley, N. J.), where PEGylated Roferon is known as Pegasys (Hoffman LaRoche); interferon alpha 2b (Intron, Schering-Plough, Madison, N. J.), where PEGylated Intron is known as PEG-Intron (Schering- Plough); interferon alpha-2c (Berofor Alpha, Boeliringer Ingelheim, Ingelheim, Germany); and consensus interferon (CIFN) as defined by determination of a consensus sequence of naturally occurring interferon alphas (Infergen®, InterMune, Inc., Brisbane, Calif), where PEGylated Infergen is referred to as PEG-Infergen.
[0081] In many embodiments, the PEG is a monomethoxyPEG molecule that reacts with primary amine groups on the IFN-α polypeptide. Methods of modifying polypeptides with monomethoxy PEG via reductive alkylation are known in the art. See, e.g., Chamow et al. (1994) Bioconj. Chem. 5:133-140.
[0082] In one non-limiting example, PEG is linked to IFN-α via an SPA linking group. SPA esters of PEG, and methods for making same, are described in U.S. Patent No. 5,672,662. SPA linkages provide for linkage to free amine groups on the IFN-α polypeptide.
[0083] For example, a PEG molecule is covalently attached via a linkage that comprises an amide bond between a propionyl group of the PEG moiety and the epsilon amino group of a surface-exposed lysine residue in the IFN-α polypeptide. Such a bond can be formed, e.g., by condensation of an α-methoxy, omega propanoic acid activated ester of PEG (mPEGspa).
[0084] As one non-limiting example, one monopegylated CIFN conjugate preferred for use herein has a linear PEG moiety of about 30 kD attached via a covalent linkage to the CIFN polypeptide, where the covalent linkage is an amide bond between a propionyl group of the PEG moiety and the epsilon amino group of a surface-exposed lysine residue in the CIFN polypeptide, where the surface-exposed lysine residue is chosen from lys , lys , lys , lys , lys
Figure imgf000019_0001
, and the amide bond is formed by condensation of an α- methoxy, omega propanoic acid activated ester of PEG. Polyethylene glycol
[0085] Polyethylene glycol suitable for conjugation to an IFN-α polypeptide is soluble in water at room temperature, and has the general formula R(O-CH2-CH2)nO-R, where R is hydrogen or a protective group such as an alkyl or an alkanol group, and where n is an integer from 1 to 1000. Where R is a protective group, it generally has from 1 to 8 carbons.
[0086] In many embodiments, PEG has at least one hydroxyl group, e.g., a terminal hydroxyl group, which hydroxyl group is modified to generate a functional group that is reactive with an amino group, e.g., an epsilon amino group of a lysine residue, a free amino group at the N- terminus of a polypeptide, or any other amino group such as an amino group of asparagine, glutamine, arginine, or histidine.
[0087] In other embodiments, PEG is derivatized so that it is reactive with free carboxyl groups in the IFN-α polypeptide, e.g., the free carboxyl group at the carboxyl terminus of the IFN-α polypeptide. Suitable derivatives of PEG that are reactive with the free carboxyl group at the carboxyl-terminus of IFN-α include, but are not limited to PEG-amine, and hydrazine derivatives of PEG (e.g., PEG-NH-NH2).
[0088] In other embodiments, PEG is derivatized such that it comprises a terminal thiocarboxylic acid group, -COSH, which selectively reacts with amino groups to generate amide derivatives. Because of the reactive nature of the thio acid, selectivity of certain amino groups over others is achieved. For example, -SH exhibits sufficient leaving group ability in reaction with N-terminal amino group at appropriate pH conditions such that the ε-amino groups in lysine residues are protonated and remain non-nucleophilic. On the other hand, reactions under suitable pH conditions may make some of the accessible lysine residues to react with selectivity.
[0089] In other embodiments, the PEG comprises a reactive ester such as an N-hydroxy succinimidate at the end of the PEG chain. Such an N-hydroxysuccinimidate-containing PEG molecule reacts with select amino groups at particular pH conditions such as neutral 6.5-7.5. For example, the N-terminal amino groups may be selectively modified under neutral pH conditions. However, if the reactivity of the reagent were extreme, accessible-NH2 groups of lysine may also react.
[0090] The PEG can be conjugated directly to the IFN-α polypeptide, or through a linker. In some embodiments, a linker is added to the IFN-α polypeptide, forming a linker-modified IFN- α polypeptide. Such linkers provide various functionalities, e.g., reactive groups such sulfhydryl, amino, or carboxyl groups to couple a PEG reagent to the linker-modified IFN-α polypeptide.
[0091] In some embodiments, the PEG conjugated to the IFN-α polypeptide is linear. In other embodiments, the PEG conjugated to the IFN-α polypeptide is branched. Branched PEG derivatives such as those described in U.S. Pat. No. 5,643,575, "star-PEG's" and multi-armed PEG's such as those described in Shearwater Polymers, Inc. catalog "Polyethylene Glycol Derivatives 1997-1998." Star PEGs are described in the art including, e.g., in U.S. Patent No. 6,046,305.
[0092] PEG having a molecular weight in a range of from about 2 kDa to about 100 kDa, is generally used, where the term "about," in the context of PEG, indicates that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight. For example, PEG suitable for conjugation to IFN-α has a molecular weight of from about 2 kDa to about 5 kDa, from about 5 kDa to about 10 kDa, from about 10 kDa to about 15 IcDa, from about 15 kDa to about 20 kDa, from about 20 kDa to about 25 kDa, from about 25 IcDa to about 30 kDa, from about 30 kDa to about 40 IcDa, from about 40 kDa to about 50 IcDa, from about 50 kDa to about 60 kDa, from about 60 kDa to about 70 kDa, from about 70 IcDa to about 80 kDa, from about 80 kDa to about 90 kDa, or from about 90 kDa to about 100 kDa. Preparing PEG-IFN-α conjugates
[0093] As discussed above, the PEG moiety can be attached, directly or via a linker, to an amino acid residue at or near the N-terminus, internally, or at or near the C-terminus of the IFN-α polypeptide. Conjugation can be carried out in solution or in the solid phase. N-terminal linkage
[0094] Methods for attaching a PEG moiety to an amino acid residue at or near the N-terminus of an IFN-α polypeptide are known in the art. See, e.g., U.S. Patent No. 5,985,265.
[0095] In some embodiments, known methods for selectively obtaining an N-terminally chemically modified IFN-α are used. For example, a method of protein modification by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminus) available for derivatization in a particular protein can be used. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved. The reaction is performed at pH which allows one to take advantage of the pKa differences between the ε- amino groups of the lysine residues and that of the α-amino group of the N-terminal residue of the protein. By such selective derivatization attachment of a PEG moiety to the IFN-α is controlled: the conjugation with the polymer takes place predominantly at the N-terminus of the IFN-α and no significant modification of other reactive groups, such as the lysine side chain amino groups, occurs. C-terminal linkage
[0096] N-terminal-specific coupling procedures such as described in U.S. Patent No. 5,985,265 provide predominantly monoPEGylated products. However, the purification procedures aimed at removing the excess reagents and minor multiply PEGylated products remove the N-terminal blocked polypeptides. In terms of therapy, such processes lead to significant increases in manufacturing costs. For example, examination of the structure of the well-characterized Infergen® Alfacon-1 CIFN polypeptide amino acid sequence reveals that the clipping is approximate 5% at the carboxyl terminus and thus there is only one major C- terminal sequence. Thus, in some embodiments, N-terminally PEGylated IFN-α is not used; instead, the IFN-α polypeptide is C-terminally PEGylated.
[0097] An effective synthetic as well as therapeutic approach to obtain mono PEGylated Infergen product is therefore envisioned as follows: [0098] A PEG reagent that is selective for the C-terminal can be prepared wit or without spacers. For example, polyethylene glycol modified as methyl ether at one end and having an amino function at the other end may be used as the starting material.
[0099] Preparing or obtaining a water-soluble carbodiimide as the condensing agent can be carried out. Coupling IFN-α (e.g., Infergen® Alfacon-1 CIFN or consensus interferon) with a water-soluble carbodiimide as the condensing reagent is generally carried out in aqueous medium with a suitable buffer system at an optimal pH to effect the amide linkage. A high molecular weight PEG can be added to the protein covalently to increase the molecular weight.
[00100] The reagents selected will depend on process optimization studies. A non-limiting example of a suitable reagent is ED AC or l-ethyl-3- (3-dimethylaminopropyl) carbodiimide. The water solubility of ED AC allows for direct addition to a reaction without the need for prior organic solvent dissolution. Excess reagent and the isourea formed as the by-product of the cross-linking reaction are both water-soluble and may easily be removed by dialysis or gel filtration. A concentrated solution of ED AC in water is prepared to facilitate the addition of a small molar amount to the reaction. The stock solution is prepared and used immediately in view of the water labile nature of the reagent. Most of the synthetic protocols in literature suggest the optimal reaction medium to be in pH range between 4.7 and 6.0. However the condensation reactions do proceed without significant losses in yields up to pH 7.5. Water may be used as solvent. In view of the contemplated use of Infergen, preferably the medium will be 2-(N-morpholino)efhane sulfonic acid buffer pre-titrated to pH between 4.7 and 6.0. However, 0.1M phosphate in the pH 7-7.5 may also be used in view of the fact that the product is in the same buffer. The ratios of PEG amine to the IFΝ-α molecule is optimized such that the C- terminal carboxyl residue(s) are selectively PEGylated to yield monoPEGylated derivative(s).
[00101] Even though the use of PEG amine has been mentioned above by name or structure, such derivatives are meant to be exemplary only, and other groups such as hydrazine derivatives as in PEG-NH-NH2 which will also condense with the carboxyl group of the IFN-α protein, can also be used. In addition to aqueous phase, the reactions can also be conducted on solid phase. Polyethylene glycol can be selected from list of compounds of molecular weight ranging from 300-40000. The choice of the various polyethylene glycols will also be dictated by the coupling efficiency and the biological performance of the purified derivative in vitro and in vivo i.e., circulation times, anti viral activities etc.
[00102] Additionally, suitable spacers can be added to the C-terminal of the protein. The spacers may have reactive groups such as SH, NH2 or COOH to couple with appropriate PEG reagent to provide the high molecular weight IFN-α derivatives. A combined solid/solution phase methodology can be devised for the preparation of C-terminal pegylated interferons. For example, the C-terminus of IFN-α is extended on a solid phase using a Gly-Gly-Cys-NH2 spacer and then monopegylated in solution using activated dithiopyridyl-PEG reagent of appropriate molecular weights. Since the coupling at the C-terminus is independent of the blocking at the N-terminus, the envisioned processes and products will be beneficial with respect to cost (a third of the protein is not wasted as in N-terminal PEGylation methods) and contribute to the economy of the therapy to treat chronic hepatitis C infections, liver fibrosis etc.
[00103] There may be a more reactive carboxyl group of amino acid residues elsewhere in the molecule to react with the PEG reagent and lead to monoPEGylation at that site or lead to multiple PEGylations in addition to the -COOH group at the C-terminus of the IFN-α. It is envisioned that these reactions will be minimal at best owing to the steric freedom at the C- terminal end of the molecule and the steric hindrance imposed by the carbodiimides and the PEG reagents such as in branched chain molecules. It is therefore the preferred mode of PEG modification for Infergen and similar such proteins, native or expressed in a host system, which may have blocked N-termini to varying degrees to improve efficiencies and maintain higher in vivo biological activity.
[00104] Another method of achieving C-terminal PEGylation is as follows. Selectivity of C- terminal PEGylation is achieved with a sterically hindered reagent which excludes reactions at carboxyl residues either buried in the helices or internally in IFN-α. For example, one such reagent could be a branched chain PEG ~40kd in molecular weight and this agent could be synthesized as follows:
[00105] OH3C-(CH2CH2O)n-CH2CH2NH2 + Glutamic Acid i.e., HOCO-CH2CH2CH(ΝH2 COOH is condensed with a suitable agent e.g., dicyclohexyl carbodiimide or water-soluble ED AC to provide the branched chain PEG agent OH3C-(CH2CH2O)n- CH2CH2NHCOCH(NH2)CH2OCH3-(CH2CH2O)n-CH2CH2NHCOCH2.
O II H3C-0-(CH2CH20)n-CH2CH2NH2+ HO C-CH2CH2CH-C00H
CHNH2 RDAC
H3C-0-(CH2CH20)n-CH2CH2NH-CO
CHNH2 (CH2)2 H3C-0-(CH2CH2θ)n-CH2CH2NH-CO
[00106] This reagent can be used in excess to couple the amino group with the free and flexible carboxyl group of IFN-α to form the peptide bond.
[00107] If desired, PEGylated IFN-α is separated from unPEGylated IFN-α using any known method, including, but not limited to, ion exchange chromatography, size exclusion chromatography, and combinations thereof. For example, where the PEG-IFN-α conjugate is a monoPEGylated IFN-α, the products are first separated by ion exchange cl romatography to obtain material having a charge characteristic of monoPEGylated material (other multi- PEGylated material having the same apparent charge may be present), and then the monoPEGylated materials are separated using size exclusion cliromatography. IFN-β
[00108] The term interferon-beta ("IFN-β") includes IFN-β polypeptides that are naturally occurring; non-naturally-occurring IFN-β polypeptides; and analogs of naturally occurring or non-naturally occurring IFN-β that retain antiviral activity of a parent naturally-occurring or non-naturally occurring IFN-β.
[00109] Any of a variety of beta interferons can be administered in conjunction with a subject method. Suitable beta interferons include, but are not limited to, naturally-occurring IFN-β; IFN-βla, e.g., Avonex® (Biogen, Inc.), and Rebif® (Serono, SA); IFN-βlb (Betaseron®; Berlex); and the like.
[00110] The IFN-β formulation may comprise an N-blocked species, wherein the N-terminal amino acid is acylated with an acyl group, such as a formyl group, an acetyl group, a malonyl group, and the like. Also suitable for use is a consensus IFN-β.
[00111] IFN-β polypeptides can be produced by any known method. DNA sequences encoding IFN-β may be synthesized using standard methods. In many embodiments, IFN-β polypeptides are the products of expression of manufactured DNA sequences transformed or transfected into bacterial hosts, e.g., E. coli, or in eukaryotic host cells (e.g., yeast; mammalian cells, such as CHO cells; and the like). In these embodiments, the IFN-β is "recombinant IFN- β." Where the host cell is a bacterial host cell, the IFN-β is modified to comprise an N- terminal methionine.
[00112] It is to be understood that IFN-β as described herein may comprise one or more modified amino acid residues, e.g., glycosylations, chemical modifications, and the like. IFN-tau
[00113] The tenri interferon-tau includes IFN-tau polypeptides that are naturally occurring; non- naturally-occurring IFN-tau polypeptides; and analogs of naturally occurring or non-naturally occurring IFN-tau that retain antiviral activity of a parent naturally-occurring or non-naturally occurring IFN-tau.
[00114] Suitable tau interferons include, but are not limited to, naturally-occurring IFN-tau; Tauferon® (Pepgen Corp.); and the like.
[00115] IFN-tau may comprise an amino acid sequence as set forth in any one of GenBank Accession Nos. P15696; P56828; P56832; P56829; P56831; Q29429; Q28595; Q28594; S08072; Q08071; Q08070; Q08053; P56830; P28169; P28172; and P28171. The sequence of any known IFN-tau polypeptide may be altered in various ways known in the art to generate targeted changes in sequence. A variant polypeptide will usually be substantially similar to the sequences provided herein, i.e. will differ by at least one amino acid, and may differ by at least two but not more than about ten amino acids. The sequence changes may be substitutions, insertions or deletions. Conservative amino acid substitutions typically include substitutions within the following groups: (glycine, alanine); (valine, isoleucine, leucine); (aspartic acid, glutamic acid); (asparagine, glutamine); (serine, threonine); (lysine, arginine); or (phenylalanine, tyrosine).
[00116] Modifications of interest that may or may not alter the primary amino acid sequence include chemical derivatization of polypeptides, e.g., acetylation, or carboxylation; changes in amino acid sequence that introduce or remove a glycosylation site; changes in amino acid sequence that make the protein susceptible to PEGylation; and the like. Also included are modifications of glycosylation, e.g. those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g. by exposing the polypeptide to enzymes that affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes. Also embraced are sequences that have phosphorylated amino acid residues, e.g. phosphotyrosine, phosphoserine, or phosphothreonine. [00117] The IFN-tau fonnulation may comprise an N-blocked species, wherein the N-terminal amino acid is acylated with an acyl group, such as a formyl group, an acetyl group, a malonyl group, and the like. Also suitable for use is a consensus IFN-tau.
[00118] IFN-tau polypeptides can be produced by any known method. DNA sequences encoding IFN-tau may be synthesized using standard methods. In many embodiments, IFN-tau polypeptides are the products of expression of manufactured DNA sequences transformed or transfected into bacterial hosts, e.g., E. coli, or in eukaryotic host cells (e.g., yeast; mammalian cells, such as CHO cells; and the like). In these embodiments, the IFN-tau is "recombinant IFΝ-tau." Where the host cell is a bacterial host cell, the IFN-tau is modified to comprise an N-terminal methionine.
[00119] It is to be understood that IFN-tau as described herein may comprise one or more modified amino acid residues, e.g., glycosylations, chemical modifications, and the like. IFN-ω
[00120] The term interferon-omega ("IFΝ-ω") includes IFN-ω polypeptides that are naturally occurring; non-naturally-occurring IFN-ω polypeptides; and analogs of naturally occurring or non-naturally occurring IFN-ω that retain antiviral activity of a parent naturally-occurring or non-naturally occurring IFN-ω.
[00121] Any known omega interferon can be administered in a subject method. Suitable IFN-ω include, but are not limited to, naturally-occurring IFN-ω; recombinant IFN-ω, e.g., Biomed 510 (BioMedicines); and the like.
[00122] IFN-ω may comprise an amino acid sequence as set forth in GenBank Accession No. NP_002168; or AAA70091. The sequence of any known IFN-ω polypeptide may be altered in various ways known in the art to generate targeted changes in sequence. A variant polypeptide will usually be substantially similar to the sequences provided herein, i.e. will differ by at least one amino acid, and may differ by at least two but not more than about ten amino acids. The sequence changes may be substitutions, insertions or deletions. Conservative amino acid substitutions typically include substitutions within the following groups: (glycine, alanine); (valine, isoleucine, leucine); (aspartic acid, glutamic acid); (asparagine, glutamine); (serine, fhreonine); (lysine, arginine); or (phenylalanine, tyrosine).
[00123] Modifications of interest that may or may not alter the primary amino acid sequence include chemical derivatization of polypeptides, e.g., acetylation, or carboxylation; changes in amino acid sequence that introduce or remove a glycosylation site; changes in amino acid sequence that make the protein susceptible to PEGylation; and the like. Also included are modifications of glycosylation, e.g. those made by modifying the'glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g. by exposing the polypeptide to enzymes that affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes. Also embraced are sequences that have phosphorylated amino acid residues, e.g. phosphotyrosine, phosphoserine, or phosphothreonine.
[00124] The IFN-ω formulation may comprise an N-bloclced species, wherein the N-terminal amino acid is acylated with an acyl group, such as a formyl group, an acetyl group, a malonyl group, and the like. Also suitable for use is a consensus IFN-ω.
[00125] IFN-ω polypeptides can be produced by any known method. DNA sequences encoding IFN-ω may be synthesized using standard methods. In many embodiments, IFN-ω polypeptides are the products of expression of manufactured DNA sequences transformed or transfected into bacterial hosts, e.g., E. coli, or in eukaryotic host cells (e.g., yeast; mammalian cells, such as CHO cells; and the like). In these embodiments, the IFN-ω is "recombinant IFN- ω." Where the host cell is a bacterial host cell, the IFN-ω is modified to comprise an N- terminal methionine.
[00126] It is to be understood that IFN-ω as described herein may comprise one or more modified amino acid residues, e.g., glycosylations, chemical modifications, and the like. Type II Interferon receptor agonists
[00127] Type II interferon receptor agonists include any naturally occurring or non-naturally- occurring ligand of a human Type II interferon receptor that binds to and causes signal transduction via the receptor. Type II interferon receptor agonists include interferons, including naturally-occurring interferons, modified interferons, synthetic interferons, pegylated interferons, fusion proteins comprising an interferon and a heterologous protein, shuffled interferons; antibody specific for an interferon receptor; non-peptide chemical agonists; and the like.
[00128] A specific example of a Type II interferon receptor agonist is IFN-gamma and variants thereof. While the present invention exemplifies use of an IFN-gamma polypeptide, it will be readily apparent that any Type II interferon receptor agonist can be used in a subject method. Interferon-Gamma
[00129] The nucleic acid sequences encoding IFN-gamma polypeptides may be accessed from public databases, e.g., Genbank, journal publications, and the like. While various mammalian IFN-gamma polypeptides are of interest, for the treatment of human disease, generally the human protein will be used. Human IFN-gamma coding sequence may be found in Genbank, accession numbers X13274; V00543; andNM_000619. The corresponding genomic sequence may be found in Genbank, accession numbers J00219; M37265; and V00536. See, for example. Gray et al. (1982) Nature 295:501 (Genbank X13274); and Rinderknecht et al. (1984) J.B.C 259:6790.
[00130] IFN-γlb (Actimmune®; human interferon) is a single-chain polypeptide of 140 amino acids. It is made recombinantly in E.coli and is unglycosylated (Rinderknecht et al. 1984, J Biol. Chem. 259:6790-6797). Recombinant IFN-gamma as discussed in U.S. Patent No. 6,497,871 is also suitable for use herein.
[00131] The IFN-gamma to be used in the methods of the present invention may be any of natural IFN-gamma, recombinant IFN-gamma and the derivatives thereof so far as they have an IFN-γ activity, particularly human IFN-gamma activity. Human IFN-gamma exhibits the antiviral and anti-proliferative properties characteristic of the interferons, as well as a number of other immunomodulatory activities, as is known in the art. Although IFN-gamma is based on the sequences as provided above, the production of the protein and proteolytic processing can result in processing variants thereof. The unprocessed sequence provided by Gray et al., supra, consists of 166 amino acids (aa). Although the recombinant IFN-gamma produced in E. coli was originally believed to be 146 amino acids, (commencing at amino acid 20) it was subsequently found that native human IFN-gamma is cleaved after residue 23, to produce a 143 aa protein, or 144 aa if the terminal methionine is present, as required for expression in bacteria. During purification, the mature protein can additionally be cleaved at the C terminus after reside 162 (referring to the Gray et al. sequence), resulting in a protein of 139 amino acids, or 140 amino acids if the initial methionine is present, e.g. if required for bacterial expression. The N-terminal methionine is an artifact encoded by the mRNA translational "start" signal AUG that, in the particular case of E. coli expression is not processed away. In other microbial systems or eukaryotic expression systems, methionine may be removed.
[00132] For use in the subject methods, any of the native IFN-gamma peptides, modifications and variants thereof, or a combination of one or more peptides may be used. IIN-gamma peptides of interest include fragments, and can be variously truncated at the carboxyl terminus relative to the full sequence. Such fragments continue to exhibit the characteristic properties of human gamma interferon, so long as amino acids 24 to about 149 (numbering from the residues of the unprocessed polypeptide) are present. Extraneous sequences can be substituted for the amino acid sequence following amino acid 155 without loss of activity. See, for example, U.S. Patent No. 5,690,925. Native IFN- gamma moieties include molecules variously extending from amino acid residues 24-150; 24-151, 24-152; 24- 153, 24-155; and 24-157. Any of these variants, and other variants known in the art and having IFN-γ activity, may be used in the present methods. [00133] The sequence of the IFN-γ polypeptide may be altered in various ways known in the art to generate targeted changes in sequence. A variant polypeptide will usually be substantially similar to the sequences provided herein, i.e., will differ by at least one amino acid, and may differ by at least two but not more than about ten amino acids. The sequence changes may be substitutions, insertions or deletions. Scanning mutations that systematically introduce alanine, or other residues, may be used to determine key amino acids. Specific amino acid substitutions of interest include conservative and non-conservative changes. Conservative amino acid substitutions typically include substitutions within the following groups: (glycine, alanine); (valine, isoleucine, leucine); (aspartic acid, glutamic acid); (asparagine, glutamine); (serine, threonine); (lysine, arginine); or (phenylalanine, tyrosine).
[00134] Modifications of interest that may or may not alter the primary amino acid sequence include chemical derivatization of polypeptides, e.g., acetylation, or carboxylation; changes in amino acid sequence that introduce or remove a glycosylation site; changes in amino acid sequence that make the protein susceptible to PEGylation; and the like. IFN-gamma may be modified with one or more polyethylene glycol moieties (PEGylated). In one embodiment, the invention contemplates the use of IFN-gamma variants with one or more non-naturally occurring glycosylation and/or pegylation sites that are engineered to provide glycosyl- and/or PEG-derivatized polypeptides with reduced serum clearance, such as the IFN-gamma polypeptide variants described in International Patent Publication No. WO 01/36001. Also included are modifications of glycosylation, e.g., those made by modifying the glycosylation patterns of a polypeptide during its synthesis and processing or in further processing steps; e.g., by exposing the polypeptide to enzymes that affect glycosylation, such as mammalian glycosylating or deglycosylating enzymes. Also embraced are sequences that have phosphorylated amino acid residues, e.g., phosphotyrosine, phosphoserine, or phosphothreonine.
[00135] Included in the subject invention is the use of polypeptides that have been modified using ordinary chemical techniques so as to improve their resistance to proteolytic degradation, to optimize solubility properties, or to render them more suitable as a therapeutic agent. For examples, the backbone of the peptide may be cyclized to enhance stability (see, for example, Friedler et al. 2000, J Biol. Chem. 275:23783-23789). Analogs may be used that include residues other than naturally occurring L-amino acids, e.g., D-amino acids or non-naturally occurring synthetic amino acids. The protein may be pegylated to enhance stability.
[00136] The polypeptides may be prepared by in vitro synthesis, using conventional methods as known in the art, by recombinant methods, or may be isolated from cells induced or naturally producing the protein. The particular sequence and the manner of preparation will be determined by convenience, economics, purity required, and the like. If desired, various groups may be introduced into the polypeptide during synthesis or during expression, which allow for linking to other molecules or to a surface. Thus cysteines can be used to make thioethers, histidines for linking to a metal ion complex, carboxyl groups for forming amides or esters, amino groups for forming amides, and the like.
[00137] The polypeptides may also be isolated and purified in accordance with conventional methods of recombinant synthesis. A lysate may be prepared of the expression host and the lysate purified using HPLC, exclusion chromatography, gel electrophoresis, affinity chromatography, or other purification technique. For the most part, the compositions which are used will comprise at least 20% by weight of the desired product, more usually at least about 15% by weight, preferably at least about 95%) by weight, and for therapeutic purposes, usually at least about 99.5%> by weight, in relation to contaminants related to the method of preparation of the product and its purification. Usually, the percentages will be based upon total protein. TNF Antagonists
[00138] Suitable TNF-α antagonists for use herein include agents that decrease the level of TNF-α synthesis, agents that block or inhibit the binding of TNF-α to a TNF-α receptor (TNFR), and agents that block or inhibit TNFR-mediated signal transduction. Unless otherwise expressly stated, every reference to a "TNF-α antagonist" or "TΝF antagonist" herein will be understood to mean a TTSfF-α antagonist other than pirfenidone or a pirfenidone analog.
[00139] As used herein, the terms "TNF receptor polypeptide" and "TΝFR polypeptide" refer to polypeptides derived from TNFR (from any species) which are capable of binding TNF. Two distinct cell-surface TNFRs have described: Type II TΝFR (or p75 TNFR or TΝFRII) and Type I TΝFR (or p55 TΝFR or TΝFRI). The mature full-length human p75 TΝFR is a glycoprotein having a molecular weight of about 75-80 kilodaltons (kD). The mature full- length human p55 TNFR is a glycoprotein having a molecular weight of about 55-60 kD. Exemplary TNFR polypeptides are derived from TNFR Type I and/or TNFR type II. Soluble TNFR includes p75 TNFR polypeptide; fusions of p75 TNFR with heterologous fusion partners, e.g., the Fc portion of an immunoglobulin.
[00140] TNFR polypeptide may be an intact TNFR or a suitable fragment of TΝFR. U.S. Pat. No. 5,605,690 provides examples of TNFR polypeptides, including soluble TΝFR polypeptides, appropriate for use in the present invention. In many embodiments, the TΝFR polypeptide comprises an extracellular domain of TNFR. In some embodiments, the TNFR polypeptide is a fusion polypeptide comprising an extracellular domain of TNFR linked to a constant domain of an immunoglobulin molecule, hi other embodiments, the TΝFR polypeptide is a fusion polypeptide comprising an extracellular domain of the p75 TΝFR linked to a constant domain of an IgGl molecule. In some embodiments, when administration to humans is contemplated, an Ig used for fusion proteins is human, e.g., human IgGl .
[00141] Monovalent and multivalent fonns of TΝFR polypeptides may be used in the present invention. Multivalent forms of TNFR polypeptides possess more than one TNF binding site. In some embodiments, the TΝFR is a bivalent, or dimeric, form of TNFR. For example, as described in U.S. Pat. No. 5,605,690 and in Mohler et al, 1993, J. Immunol., 151:1548-1561, a chimeric antibody polypeptide with TNFR extracellular domains substituted for the variable domains of either or both of the immunoglobulin heavy or light chains would provide a TNFR polypeptide for the present invention. Generally, when such a chimeric TNFR: antibody polypeptide is produced by cells, it forms a bivalent molecule through disulfide linkages between the immunoglobulin domains. Such a chimeric TΝFR: antibody polypeptide is referred to as TΝFR:Fc.
[00142] In one embodiment, a subject method involves administration of an effective amount of the soluble TNFR ENBREL®. ENBREL® is a dimeric fusion protein consisting of the extracellular ligand-binding portion of the human 75 kilodalton (pi 5) TNFR linked to the Fc portion of human IgGl. The Fc component of ENBREL® contains the CH2 domain, the CH3 domain and hinge region, but not the CHI domain of IgGl. ENBREL® is produced in a Chinese hamster ovary (CHO) mammalian cell expression system. It consists of 934 amino acids and has an apparent molecular weight of approximately 150 kilodaltons. Smith et al. (1990) Science 248:1019-1023; Mohler et al. (1993) J. Immunol. 151 :1548-1561; U.S. Pat No. 5,395,760; and U.S. Pat. No. 5,605,690.
[00143] Also suitable for use are monoclonal antibodies that bind TNF-α. Monoclonal antibodies include "humanized" mouse monoclonal antibodies; chimeric antibodies; monoclonal antibodies that are at least about 80%), at least about 90%, at least about 95%, or 100% human in amino acid sequence; and the like. See, e.g., WO 90/10077; WO 90/04036; and WO 92/02190. Suitable monoclonal antibodies include antibody fragments, such as Fv, F(ab')2 and Fab; synthetic antibodies; artificial antibodies; phage display antibodies; and the like.
[00144] Examples of suitable monoclonal antibodies include Infliximab (REMICADE®, Centocor); and Adalimumab (HUMIRA™, Abbott) REMICADE® is a chimeric monoclonal anti-TNF-α antibody that includes about 25% mouse amino acid sequence and about 75% human amino acid sequence. REMICADE® comprises a variable region of a mouse monoclonal anti-TNF-α antibody fused to the constant region of a human IgGl . Elliott et al. (1993) Arthritis Rheum. 36:1681-1690; Elliott et al. (1994) Lancet 344:1105-1110; Baert et al. (1999) Gastroenterology 116:22-28. HUMIRA™ is a human, full-length IgGl monoclonal antibody that was identified using phage display technology. Piascik (2003) J Am. Pharm. Assoc. 43:327-328.
[00145] Also included in the term "TNF antagonist," and therefore suitable for use in a subject method, are stress-activated protein kinase (SAPK) inhibitors. SAPK inhibitors are known in the art, and include, but are not limited to 2-alkyl imidazoles disclosed in U.S. Patent No. 6,548,520; 1,4,5-substituted imidazole compounds disclosed in U.S. Patent No. 6,489,325; 1,4,5-substituted imidazole compounds disclosed in U.S. Patent No. 6,569,871; heteroaryl aminophenyl ketone compounds disclosed in Published U.S. Patent Application No. 2003/0073832; pyridyl imidazole compounds disclosed in U.S. Patent No. 6,288,089; and heteroaryl aminobenzophenones disclosed in U.S. Patent No. 6,432,962. Also of interest are compounds disclosed in U.S. Patent Application Publication No. 2003/0149041 ; and U.S. Patent No. 6,214,854. A stress-activated protein kinase is a member of a family of mitogen- activated protein kinases which are activated in response to stress stimuli. SAPK include, but are not limited to, p38 (Lee et al. (1994) Nature 372:739) and c-jun N-terminal kinase (JNK).
[00146] Methods to assess TNF antagonist activity are known in the art and exemplified herein. For example, TNF antagonist activity may be assessed with a cell-based competitive binding assay. In such an assay, radiolabeled TNF is mixed with serially diluted TNF antagonist and cells expressing cell membrane bound TNFR. Portions of the suspension are centrifuged to separate free and bound TNF and the amount of radioactivity in the free and bound fractions determined. TNF antagonist activity is assessed by inhibition of TNF binding to the cells in the presence of the TNF antagonist.
[00147] As another example, TNF antagonists may be analyzed for the ability to neutralize TNF activity in vitro in a bioassay using cells susceptible to the cytotoxic activity of TNF as target cells. In such an assay, target cells, cultured with TNF, are treated with varying amounts of TNF antagonist and subsequently are examined for cytolysis. TNF antagonist activity is assessed by a decrease in TNF-induced target cell cytolysis in the presence of the TNF antagonist. Pirfenidone and Analogs Thereof [00148] Pirfenidone (5 -methyl- l-phenyl-2-(lH)-pyridone) and pirfenidone analogs are used in certain combination therapies of the invention. Pirfenidone
Figure imgf000033_0001
Pirfenidone analogs
Figure imgf000033_0002
II.A ILB
Figure imgf000033_0003
Descriptions for Substituents Ri, R2, X
[00149] Ri: carbocyclic (saturated and unsaturated), heterocyclic (saturated or unsaturated), alkyls (saturated and unsaturated). Examples include phenyl, benzyl, pyrimidyl, naphthyl, indolyl, pyrrolyl, furyl, thienyl, imidazolyl, cyclohexyl, piperidyl, pyrrolidyl, morpholinyl, cyclohexenyl, butadienyl, and the like.
[00150] Ri can further include substitutions on the carbocyclic or heterocyclic moieties with substituents such as halogen, nitro, amino, hydroxyl, alkoxy, carboxyl, cyano, thio, alkyl, aryl, heteroalkyl, heteroaryl and combinations thereof, for example, 4-nitrophenyl, 3-chloroρhenyl, 2,5-dinitrophenyl, 4-methoxyphenyl, 5-methyl-pyrrolyl, 2, 5-dichlorocyclohexyl, guanidinyl- cyclohexenyl and the like.
[00151] R2: alkyl, carbocylic, aryl, heterocyclic. Examples include: methyl, ethyl, propyl, isopropyl, phenyl, 4-nitrophenyl, thienyl and the like.
[00152] X: may be any number (from 1 to 3) of substituents on the carbocyclic or heterocyclic ring. The substituents can be the same or different. Substituents can include hydrogen, alkyl, heteroalkyl, aryl, heteroaryl, halo, nitro, carboxyl, hydroxyl, cyano, amino, thio, alkylamino, haloaryl and the like. [00153] The substituents may be optionally further substituted with 1-3 substituents from the group consisting of alkyl, aryl, nitro, alkoxy, hydroxyl and halo groups. Examples include: methyl, 2,3-dimethyl, phenyl, p-tolyl, 4-chlorophenyl, 4-nitrophenyl, 2,5-dichlorophenyl, furyl, thienyl and the like. [00154] Specific Examples include those shown in Table 1 :
Figure imgf000034_0001
[00155] U.S. Pat. Nos. 3,974,281; 3,839,346; 4,042,699; 4,052,509; 5,310,562; 5,518,729; 5,716,632; and 6,090,822 describe metiiods for the synthesis and formulation of pirfenidone and pirfenidone analogs in pharmaceutical compositions suitable for use in the methods of the present invention. Side effect management agents
[00156] In some embodiments, a subject combination therapy further includes administering a side effect management agent that reduces a side effect of a therapeutic agent, in an amount effective to reduce at least one side effect. Side effect management agents include palliative agents, and other agents for the treatment, reduction, or avoidance of a side effect caused by any therapeutic agent.
[00157] In some embodiments, a subject therapy with pirfenidone or pirfenidone analog further includes administering a side effect management agent in an amount effective to reduce at least one side effect of the pirfenidone or pirfenidone analog therapy. Side effects of pirfenidone or pirfenidone analog treatment include gastrointestinal disturbances and discomfort. Gastrointestinal disturbances include nausea, diarrhea, gastrointestinal cramping, and the like. In some embodiments, an effective amount of a side effect management agent reduces a side effect induced by treatment with a pirfenidone or pirfenidone analog by at least about 10%, at least about 20%, at least about 30%), at least about 40%, at least about 50%, at least about 60%, or more, compared to the rate of occurrence or the degree or extent of the side effect when the pirfenidone or pirfenidone analog therapy is administered without the side effect management agent.
[00158] In some embodiments, a subject therapy with a Type II interferon receptor agonist further includes administering a side effect management agent in an amount effective to reduce at least one side effect of the Type II interferon receptor agonist therapy. Side effects of Type II interferon receptor agonist treatment include, but are not limited to, fever, malaise, tachycardia, chills, headache, arthralgia, myalgia, myelosuppression, suicide ideation, platelet suppression, neutropenia, lymphocytopenia, erythrocytopenia (anemia), and anorexia. In some embodiments, an effective amount of a side effect management agent reduces a side effect induced by treatment with a Type II interferon receptor agonist by at least about 10%), at least about 20%>, at least about 30%>, at least about 40%, at least about 50%, at least about 60%>, or more, compared to the rate of occurrence or the degree or extent of the side effect when the Type II interferon receptor agonist therapy is administered without the side effect management agent. For example, if a fever is experienced with the Type II interferon receptor agonist therapy, then the body temperature of an individual treated with the Type II interferon receptor agonist therapy and side effect management agent according to the instant invention is reduced by at least 0.5 degree Fahrenheit, and in some embodiments is within the normal range, e.g., at or near 98.6 °F.
[00159] Suitable side effect management agents include agents that are effective in pain management; agents that ameliorate gastrointestinal discomfort; analgesics, anti- inflammatories, antipsychotics, antineurotics, anxiolytics, hematopoietic agents, and agents that reduce gastrointestinal discomfort. In addition, the invention contemplates the use of any compound for palliative care of patients suffering from pain or any other side effect in the course of treatment with a subject monotherapy or combination therapy. Exemplary side effect management agents include acetaminophen, ibuprofen, and other NSAIDs, H2 blockers, hematopoietic agents, and antacids.
[00160] Suitable H2 blockers (histamine type 2 receptor antagonists) that are suitable for use as a side effect management agent in a subject therapy include, but are not limited to, Cimetidine (e.g., Tagamet, Peptol, Nu-cimet, apo-cimetidine, non-cimetidine); Ranitidine (e.g., Zantac, Nu-ranit, Novo-randine, and apo-ranitidine); and Famotidine (Pepcid, Apo-Famotidine, and No vo-Famotidine) .
[00161] Suitable antacids include, but are not limited to, aluminum and magnesium hydroxide (Maalox®, Mylanta®); aluminum carbonate gel (Basajel®); aluminum hydroxide (Amphojel®, AlternaGEL®); calcium carbonate (Turns®, Titralac®); magnesium hydroxide; and sodium bicarbonate.
[00162] Suitable non-steroidal anti-inflammatory drugs (ΝSAIDs) include, but are not limited to, acetaminophen, salicylate, acetyl-salicylic acid (aspirin, diflunisal), ibuprofen, Motrin, Naprosyn, Nalfon, and Trilisate, indomethacin, glucametacme, acemetacin, sulindac, naproxen, piroxicam, diclofenac, benoxaprofen, ketoprofen, oxaprozin, etodolac, Icetorolac tromethamine, ketorolac, nabumetone, and the like, and mixtures of two or more of the foregoing.
[00163] Suitable hematopoietic agents include agents that prevent or restore depressed blood cell populations, including, but not limited to, erythropoietins, such as EPOGEN™ epoetin- alfa, granulocyte colony stimulating factors (G-CSFs), such as NEUPOGEN™ filgrastim, granulocyte-macrophage colony stimulating factors (GM-CSFs), thrombopoietins, etc. DOSAGES, FORMULATIONS, AND ROUTES OF ADMINISTRATION
[00164] A therapeutic agent (e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist) which is administered in a subject monotherapy or combination therapy is administered to individuals in a formulation with a pharmaceutically acceptable excipient(s). A wide variety of pharmaceutically acceptable excipients are known in the art and need not be discussed in detail herein. Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, A. Gennaro (2000) "Remington: The Science and Practice of Pharmacy," 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H.C. Ansel et al., eds., 7th ed., Lippincott, Williams, & Wilkins; and Handbook of Pharmaceutical Excipients (2000) A-H. Kibbe et al., eds., 3rd ed. Amer. Pharmaceutical Assoc. The terms "therapeutic agent" and "active agent" are used interchangeably herein.
[00165] The pharmaceutically acceptable excipients, such as vehicles, adjuvants, carriers or diluents, are readily available to the public. Moreover, pharmaceutically acceptable auxiliary substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are readily available to the public.
[00166] In the subject methods, an active agent (e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist) may be administered to the host using any convenient means capable of resulting in the desired therapeutic effect. Thus, the agents can be incorporated into a variety of formulations for therapeutic administration. More particularly, an active agent can be formulated into pharmaceutical compositions by combination with appropriate, pharmaceutically acceptable carriers or diluents, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants and aerosols.
[00167] As such, administration of an active agent (e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist) can be achieved in various ways, including oral, buccal, rectal, parenteral, intraperitoneal, intradermal, subcutaneous, intramuscular, transdermal, intratracheal, etc., administration. In some embodiments, two different routes of administration are used.
[00168] Subcutaneous administration of a therapeutic agent can be accomplished using standard methods and devices, e.g., needle and syringe, a subcutaneous injection port delivery system, and the like. See, e.g., U.S. Patent Nos. 3,547,119; 4,755,173; 4,531,937; 4,311,137; and 6,017,328. A combination of a subcutaneous injection port and a device for administration of an agent to a patient through the port is referred to herein as "a subcutaneous injection port delivery system." In some embodiments, subcutaneous administration is achieved by a combination of devices, e.g., bolus delivery by needle and syringe, followed by delivery using a continuous delivery system. [00169] In some embodiments, a therapeutic agent is delivered by a continuous delivery system. The term "continuous delivery system" is used interchangeably herein with "controlled delivery system" and encompasses continuous (e.g., controlled) delivery devices (e.g., pumps) in combination with catheters, injection devices, and the like, a wide variety of which are known in the art.
[00170] Mechanical or electromechanical infusion pumps can also be suitable for use with the present invention. Examples of such devices include those described in, for example, U.S. Pat. Nos. 4,692,147; 4,360,019; 4,487,603; 4,360,019; 4,725,852; 5,820,589; 5,643,207; 6,198,966; and the like. In general, the present methods of drug delivery can be accomplished using any of a variety of refillable, pump systems. Pumps provide consistent, controlled release over time. Typically, the therapeutic agent is in a liquid formulation in a drug-impermeable reservoir, and is delivered in a continuous fashion to the individual.
[00171] In one embodiment, the drug delivery system is an at least partially implantable device. The implantable device can be implanted at any suitable implantation site using methods and devices well known in the art. An implantation site is a site within the body of a subject at which a drug delivery device is introduced and positioned. Implantation sites include, but are not necessarily limited to a subdermal, subcutaneous, intramuscular, or other suitable site within a subject's body. Subcutaneous implantation sites are generally preferred because of convenience in implantation and removal of the drug delivery device.
[00172] Drug release devices suitable for use in the invention may be based on any of a variety of modes of operation. For example, the drug release device can be based upon a diffusive system, a convective system, or an erodible system (e.g., an erosion-based system). For example, the drug release device can be an electrochemical pump, osmotic pump, an electroosmotic pump, a vapor pressure pump, or osmotic bursting matrix, e.g., where the drug is incorporated into a polymer and the polymer provides for release of drug formulation concomitant with degradation of a drug-impregnated polymeric material (e.g., a biodegradable, drug-impregnated polymeric material). In other embodiments, the drug release device is based upon an electrodiffusion system, an electrolytic pump, an effervescent pump, a piezoelectric pump, a hydrolytic system, etc.
[00173] Drug release devices based upon a mechanical or electromechanical infusion pump can also be suitable for use with the present invention. Examples of such devices include those described in, for example, U.S. Pat. Nos. 4,692,147; 4,360,019; 4,487,603; 4,360,019; 4,725,852, and the like. In general, the present methods of drug delivery can be accomplished using any of a variety of refillable, non-exchangeable pump systems. Pumps and other convective systems are generally preferred due to their generally more consistent, controlled release over time. Osmotic pumps are used in some embodiments, due to their combined advantages of more consistent controlled release and relatively small size (see, e.g., PCT published application no. WO 97/27840 and U.S. Pat. Nos. 5,985,305 and 5,728,396)). Exemplary osmotically-driven devices suitable for use in the invention include, but are not necessarily limited to, those described in U.S. Pat. Nos. 3,760,984; 3,845,770; 3,916,899; 3,923,426; 3,987,790; 3,995,631; 3,916,899; 4,016,880; 4,036,228; 4,111,202; 4,111,203; 4,203,440; 4,203,442; 4,210,139; 4,327,725; 4,627,850; 4,865,845; 5,057,318; 5,059,423; 5,112,614; 5,137,727; 5,234,692; 5,234,693; 5,728,396; and the like.
[00174] In some embodiments, the drug delivery device is an implantable device. The drug delivery device can be implanted at any suitable implantation site using methods and devices well known in the art. As noted infra, an implantation site is a site within the body of a subject at which a drug delivery device is introduced and positioned. Implantation sites include, but are not necessarily limited to a subdermal, subcutaneous, intramuscular, or other suitable site within a subject's body.
[00175] In some embodiments, a therapeutic agent is delivered using an implantable drug delivery system, e.g., a system that is programmable to provide for administration of a an active agent (e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist). Exemplary programmable, implantable systems include implantable infusion pumps. Exemplary implantable infusion pumps, or devices useful in connection with such pumps, are described in, for example, U.S. Pat. Nos. 4,350,155; 5,443,450; 5,814,019; 5,976,109; 6,017,328; 6,171,276; 6,241,704; 6,464,687; 6,475,180; and 6,512,954. A further exemplary device that can be adapted for the present invention is the Syncl romed infusion pump (Medtronic).
[00176] In pharmaceutical dosage forms, an active agent (e.g., pirfenidone or pirfenidone analog; a Type I interferon receptor agonist; a Type II interferon receptor agonist; a TNF antagonist) may be administered in the form of its pharmaceutically acceptable salts, or the active agent may also be used alone or in appropriate association, as well as in combination, with other pharmaceutically active compounds. The following methods and excipients are merely exemplary and are in no way limiting.
[00177] For oral preparations, an active agent can be used alone or in combination with appropriate additives to make tablets, powders, granules or capsules, for example, with conventional additives, such as lactose, mannitol, corn starch or potato starch; with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch or gelatins; with disintegrators, such as corn starch, potato starch or sodium carboxymethylcellulose; with lubricants, such as talc or magnesium stearate; and if desired, with diluents, buffering agents, moistening agents, preservatives and flavoring agents.
[00178] The agents can be formulated into preparations for injection by dissolving, suspending or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other similar oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives.
[00179] Furthermore, the agents can be made into suppositories by mixing with a variety of bases such as emulsifying bases or water-soluble bases. The compounds of the present invention can be administered rectally via a suppository. The suppository can include vehicles such as cocoa butter, carbowaxes and polyethylene glycols, which melt at body temperature, yet are solidified at room temperature.
[00180] Unit dosage forms for oral or rectal administration such as syrups, elixirs, and suspensions may be provided wherein each dosage unit, for example, teaspoonful, tablespoonful, tablet or suppository, contains a predetermined amount of the composition containing one or more active agents. Similarly, unit dosage forms for injection or intravenous administration may comprise the active agent(s) in a composition as a solution in sterile water, normal saline or another pharmaceutically acceptable carrier.
[00181] The term "unit dosage form," as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of compounds of the present invention calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for an active agent depend on the particular agent employed and the effect to be achieved, and the pharmacodynamics associated with the agent in the host.
[00182] In connection with each of the methods described herein, the invention provides embodiments in which the therapeutic agent administered to the patient by a controlled drug delivery device. In some embodiments, the therapeutic agent is delivered to the patient substantially continuously or continuously by the controlled drug delivery device. Optionally, an implantable infusion pump is used to deliver the therapeutic agent to the patient substantially continuously or continuously by subcutaneous infusion.
[00183] In other embodiments, a therapeutic agent is administered to the patient so as to achieve and maintain a desired average daily serum concentration of the therapeutic agent at a substantially steady state for the duration of the therapy. Optionally, an implantable infusion pump is used to deliver the therapeutic agent to the patient by subcutaneous infusion so as to achieve and maintain a desired average daily serum concentration of the therapeutic agent at a substantially steady state for the duration of the therapy.
[00184] Effective dosages of pirfenidone or a specific pirfenidone analog include a weight- based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally. Other doses and formulations of pirfenidone and specific pirfenidone analogs suitable for use in the treatment of fibrotic diseases are described in U.S. Pat. Nos., 5,310,562; 5,518,729; 5,716,632; and 6,090,822.
[00185] In many embodiments, a pirfenidone or pirfenidone analog is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time. The pirfenidone or pirfenidone analog can be administered fid, bid, qd, qod, biw, tiw, qw, qow, three times per month, once monthly, substantially continuously, or continuously.
[00186] In many embodiments, multiple doses of a pirfenidone or pirfenidone analog are administered. For example, a pirfenidone or pirfenidone analog is administered once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (bid), or three times a day (tid), substantially continuously, or continuously, over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.
[00187] Those of skill in the art will readily appreciate that dose levels can vary as a function of the specific compounds, the severity of the symptoms and the susceptibility of the subject to side effects. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means. A preferred means is to measure the physiological potency of a given compound. [00188] In some embodiments, a pirfenidone or pirfenidone analog is administered in combination therapy with at least a second therapeutic agent. In some of these embodiments, a second therapeutic agent is administered throughout the entire course of pirfenidone/pirfenidone analog treatment. In other embodiments, a second therapeutic agent is administered less than the entire course of pirfenidone/pirfenidone analog treatment, e.g., only during the first phase of pirfenidone/pirfenidone analog treatment, only during the second phase of pirfenidone/pirfenidone analog treatment, or some other portion of the pirfenidone/pirfenidone analog treatment regimen.
[00189] In some embodiments, a subject combination therapy involves co-administering pirfenidone or a pirfenidone analog and a Type II interferon receptor agonist. In some of these embodiments, the Type II interferon receptor agonist is an IFN-γ.
[00190] Effective dosages of IFN-γ can range from about 0.5 μg/m2 to about 500 μg/m2, usually from about 1.5 μg/m2 to 200 μg/m2, depending on the size of the patient. This activity is based on 106 international units (U) per 50 μg of protein. IFN-γ can be administered daily, every other day, three times a week, or substantially continuously or continuously.
[00191] In specific embodiments of interest, IFN-γ is administered to an individual in a unit dosage form of from about 25 μg to about 500 μg, from about 50 μg to about 400 μg, or from about 100 μg to about 300 μg. In particular embodiments of interest, the dose is about 200 μg IFN-γ. In many embodiments of interest, IFN-γlb is administered.
[00192] Where the dosage is 200 μg IFΝ-γ per dose, the amount of IFΝ-γ per body weight (assuming a range of body weights of from about 45 kg to about 135 kg) is in the range of from about 4.4 μg IFN-γ per kg body weight to about 1.48 μg IFN-γ per kg body weight.
[00193] The body surface area of subject individuals generally ranges from about 1.33 m2 to about 2.50 m2. Thus, in many embodiments, an IFN-γ dosage ranges from about 150 μg/m2 to about 20 μg/m2. For example, an IFN-γ dosage ranges from about 20 μg/m2 to about 30 μg/m2, from about 30 μg/m2 to about 40 μg/m2, from about 40 μg/m2 to about 50 μg/m2, from about 50 μg/m2 to about 60 μg/m2, from about 60 μg/m2 to about 70 μg/m2, from about 70 μg/m2 to about 80 μg/m2, from about 80 μg/m2 to about 90 μg/m2, from about 90 μg/m2 to about 100 μg/m2, from about 100 μg/m2 to about 110 μg/m2, from about 110 μg/m2 to about 120 μg/m2, from about 120 μg/m2 to about 130 μg/m2, from about 130 μg/m2 to about 140 μg/m2, or from about 140 μg/m2 to about 150 μg/m2. In some embodiments, the dosage groups range from about 25 μg/m2 to about 100 μg/m2. In other embodiments, the dosage groups range from about 25 μg/m to about 50 μg/m . [00194] In many embodiments, IFN-γ is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time. IFN-γ can be administered tid, bid, qd, qod, biw, tiw, qw, qow, three times per month, once monthly, substantially continuously, or continuously.
[00195] In many embodiments, multiple doses of IFN-γ are administered. For example, IFN-γ is administered once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (bid), or three times a day (tid), substantially continuously, or continuously, over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.
[00196] In other embodiments, a subject combination therapy involves administering a Type I interferon receptor agonist. In some embodiments, the Type I interferon receptor agonist is an IFN-α. Effective dosages of an IFN-α range from about 3 μg to about 27 μg, from about 3 MU to about 10 MU, from about 90 μg to about 180 μg, or from about 18 μg to about 90 μg.
[00197] Effective dosages of Infergen® consensus IFN-α include about 3 μg, about 6 μg, about 9 μg, about 12 μg, about 15 μg, about 18 μg, about 21 μg, about 24 μg, about 27 μg, or about 30 μg, of drug per dose. Effective dosages of IFN-α2a and IFN-α2b range from 3 million Units (MU) to 10 MU per dose. Effective dosages of PEGASYS®PEGylated IFN-α2a contain an amount of about 90 μg to 270 μg, or about 180 μg, of drug per dose. Effective dosages of PEG-INTRON®PEGylated IFΝ-α2b contain an amount of about 0.5 μg to 3.0 μg of drug per kg of body weight per dose. Effective dosages of PEGylated consensus interferon (PEG- CIFN) contain an amount of about 18 μg to about 90 μg, or from about 27 μg to about 60 μg, or about 45 μg, of CIFN amino acid weight per dose of PEG-CIFN. Effective dosages of monoPEG (30 kD, linear)-ylated CIFN contain an amount of about 45 μg to about 270 μg, or about 60 μg to about 180 μg, or about 90 μg to about 120 μg, of drug per dose. IFN-α can be administered daily, every other day, once a week, three times a week, every other week, three times per month, once monthly, substantially continuously or continuously. [00198] In other embodiments, a subject combination therapy involves administering a TNF-α antagonist. Effective dosages of a TNF-α antagonist range from 0.1 μg to 40 mg per dose, e.g., from about 0.1 μg to about 0.5 μg per dose, from about 0.5 μg to about 1.0 μg per dose, from about 1.0 μg per dose to about 5.0 μg per dose, from about 5.0 μg to about 10 μg per dose, from about 10 μg to about 20 μg per dose, from about 20 μg per dose to about 30 μg per dose, from about 30 μg per dose to about 40 μg per dose, from about 40 μg per dose to about 50 μg per dose, from about 50 μg per dose to about 60 μg per dose, from about 60 μg per dose to about 70 μg per dose, from about 70 μg to about 80 μg per dose, from about 80 μg per dose to about 100 μg per dose, from about 100 μg to about 150 μg per dose, from about 150 μg to about 200 μg per dose, from about 200 μg per dose to about 250 μg per dose, from about 250 μg to about 300 μg per dose, from about 300 μg to about 400 μg per dose, from about 400 μg to about 500 μg per dose, from about 500 μg to about 600 μg per dose, from about 600 μg to about 700 μg per dose, from about 700 μg to about 800 μg per dose, from about 800 μg to about 900 μg per dose, from about 900 μg to about 1000 μg per dose, from about 1 mg to about 10 mg per dose, from about 10 mg to about 15 mg per dose, from about 15 mg to about 20 mg per dose, from about 20 mg to about 25 mg per dose, from about 25 mg to about 30 mg per dose, from about 30 mg to about 35 mg per dose, or from about 35 mg to about 40 mg per dose.
[00199] In some embodiments, the TNF-α antagonist is ENBREL® etanercept. Effective dosages of etanercept range from about 0.1 μg to about 40 mg per dose, from about 0.1 μg to about 1 μg per dose, from about 1 μg to about 10 μg per dose, from about 10 μg to about 100 μg per dose, from about 100 μg to about 1 mg per dose, from about 1 mg to about 5 mg per dose, from about 5 mg to about 10 mg, from about 10 mg to about 15 mg per dose, from about 15 mg to about 20 mg per dose, from about 20 mg to about 25 mg per dose, from about 25 mg to about 30 mg per dose, from about 30 mg to about 35 mg per dose, or from about 35 mg to about 40 mg per dose.
[00200] In some embodiments, effective dosages of a TNF-α antagonist are expressed as mg/kg body weight. In these embodiments, effective dosages of a TNF-α antagonist are from about 0.1 mg/kg body weight to about 10 mg/kg body weight, e.g., from about 0.1 mg/kg body weight to about 0.5 mg/kg body weight, from about 0.5 mg/kg body weight to about 1.0 mg/kg body weight, from about 1.0 mg/kg body weight to about 2.5 mg/kg body weight, from about 2.5 mg/kg body weight to about 5.0 mg/kg body weight, from about 5.0 mg/kg body weight to about 7.5 mg/kg body weight, or from about 7.5 mg/kg body weight to about 10 mg/kg body weight. [00201] In some embodiments, the TNF-α antagonist is REMICADE®. Effective dosages of REMICADE® range from about 0.1 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 0.5 mg/kg, from about 0.5 mg/kg to about 1.0 mg/kg, from about 1.0 mg/kg to about 1.5 mg/kg, from about 1.5 mg/kg to about 2.0 mg/kg, from about 2.0 mg/kg to about 2.5 mg/kg, from about 2.5 mg/kg to about 3.0 mg/kg, from about 3.0 mg/kg to about 3.5 mg/kg, from about 3.5 mg/kg to about 4.0 mg/kg, from about 4.0 mg/kg to about 4.5 mg/kg, from about 4.5 mg/kg to about 5.0 mg/kg, from about 5.0 mg/kg to about 7.5 mg/kg, or from about 7.5 mg/kg to about 10 mg/kg per dose.
[00202] In some embodiments the TNF-α antagonist is HUMIRA™. Effective dosages of HUMIRA™ range from about 0.1 μg to about 35 mg, from about- 0.1 μg to about 1 μg, from about 1 μg to about 10 μg, from about 10 μg to about 100 μg, from about 100 μg to about 1 mg, from about 1 mg to about 5 mg, from about 5 mg to about 10 mg, from about 10 mg to about 15 mg, from about 15 mg to about 20 mg, from about 20 mg to about 25 mg, from about 25 mg to about 30 mg, from about 30 mg to about 35 mg, or from about 35 mg to about 40 mg per dose.
[00203] In many embodiments, a TNF-α antagonist is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time. The TNF-α antagonist can be administered tid, bid, qd, qod, biw, tiw, qw, qow, three times per month, once monthly, once bimonthly, substantially continuously, or continuously.
[00204] In many embodiments, multiple doses of a TNF-α antagonist are administered. For example, a TNF-α antagonist is administered once bimonthly, once per month, twice per month, three times per month, every other week (qow), once per week (qw), twice per week (biw), three times per week (tiw), four times per week, five times per week, six times per week, every other day (qod), daily (qd), twice a day (bid), or three times a day (tid), substantially continuously, or continuously, over a period of time ranging from about one day to about one week, from about two weeks to about four weeks, from about one month to about two months, from about two months to about four months, from about four months to about six months, from about six months to about eight months, from about eight months to about 1 year, from about 1 year to about 2 years, or from about 2 years to about 4 years, or more.
[00205] Those of skill in the art will readily appreciate that dose levels can vary as a function of the specific compounds, the severity of the symptoms and the susceptibility of the subject to side effects. Preferred dosages for a given compound are readily determinable by those of skill in the art by a variety of means. A preferred means is to measure the physiological potency of a given compound. MONOTHERAPY AND COMBINATION THERAPY REGIMENS
[00206] In one aspect, the present invention provides pirfenidone or pirfenidone analog monotherapy for the treatment of alcoholic hepatitis. In another aspect, the present invention provides pirfenidone or pirfenidone analog combination therapy for the treatment of alcoholic hepatitis. In another aspect, the present invention provides pirfenidone or pirfenidone analog monotherapy therapy for the treatment of ALD. In another aspect, the present invention provides combination therapy for the treatment of ALD. Pirfenidone monotherapy for treating non-fibrotic alcoholic hepatitis
[00207] In one aspect, the present invention provides pirfenidone or pirfenidone analog monotherapy for the treatment of non-fibrotic alcoholic hepatitis.
[00208] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd, optionally in two or more divided doses per day, for the desired treatment duration.
[00209] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00210] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00211] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration. [00212] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00213] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00214] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00215] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00216] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog of about 1200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00217] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of non-fibrotic alcoholic hepatitis in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog of about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration. Pirfenidone combination therapy for treating non-fibrotic alcoholic hepatitis
[00218] In another aspect, the present invention provides pirfenidone or pirfenidone analog combination therapy for the treatment of non-fibrotic alcoholic hepatitis. [00219] In some embodiments, the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog and a Type II interferon receptor agonist in the treatment of non-fibrotic alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; and an effective amount of a Type II interferon receptor agonist.
[00220] In some embodiments, the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog and a TNF-α antagonist in the treatment of non-fibrotic alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; and an effective amount of a TNF-α antagonist.
[00221] In some embodiments, the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog, a Type II interferon receptor agonist and a TNF-α antagonist in the treatment of non-fibrotic alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; an effective amount of a Type II interferon receptor agonist; and an effective amount of a TNF-α antagonist.
[00222] In some embodiments, the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog, a Type II interferon receptor agonist, a Type I interferon receptor agonist and a TNF-α antagonist in the treatment of non-fibrotic alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; an effective amount of a Type II interferon receptor agonist; an effective amount of a Type I interferon receptor agonist; and an effective amount of a TNF-α antagonist.
[00223] In some embodiments, the effective amount of a Type II interferon receptor agonist referred to in any of the foregoing methods of pirfenidone combination therapy for treatment of non-fibrotic alcoholic hepatitis can be selected from a fixed dosage of IFN-γ in the range from about 25 μg to about 500 μg, from about 50 μg to about 400 μg, or from about 100 μg to about 300 μg, or about 200 μg, of IFN-γ per dose subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously for the duration of pirfenidone therapy.
[00224] In some embodiments, the effective amount of a Type I interferon receptor agonist referred to in any of the foregoing methods of pirfenidone combination therapy for treatment of non-fibrotic alcoholic hepatitis can be selected from: (1) a fixed dosage of Infergen® consensus IFN-α of about 3 μg, about 6 μg, about 9 μg, about 12 μg, about 15 μg, about 18 μg, about 21 μg, about 24 μg, about 27 μg, or about 30 μg, of drug per dose subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously for the duration of pirfenidone therapy; (2) a fixed dosage of IFN-α 2a, 2b or 2c in the range from about 3 million Units (MU) to 10 MU of drug per dose subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously for the duration of pirfenidone therapy; (3) a fixed dosage of PEGASYS® peginterferon-alfa 2a in the range of about 90 μg to 270 μg, or about 180 μg, of drug per dose subcutaneously qw, qow, three times per month, or monthly for the duration of pirfenidone therapy; (4) a weight-based dosage of PEG-LΝTRON® peginterferon- alfa 2b in the range of about 0.5 μg to 3.0 μg of drug per kg of body weight per dose subcutaneously qw, qow, three times per month, or monthly for the duration of pirfenidone therapy; (5) a fixed dosage of PEGylated consensus interferon (PEG-CIFN) in the range of about 18 μg to about 90 μg, or from about 27 μg to about 60 μg, or about 45 μg, of CIFN amino acid weight per dose of PEG-CIFN subcutaneously qw, qow, three times per month, or monthly for the duration of pirfenidone therapy; or (6) a fixed dosage of monoPEG (30 lcD, linear)-ylated CIFN in the range of about 45 μg to about 270 μg, or about 60 μg to about 180 μg, or about 90 μg to about 120 μg, of drug per dose subcutaneously qw, qow, three times per month, or monthly for the duration of pirfenidone therapy.
[00225] In some embodiments, the effective amount of a TNF-α antagonist referred to in any of the foregoing methods of pirfenidone combination therapy for treatment of non-fibrotic alcoholic hepatitis can be selected from: (1) a fixed dosage of ENBREL® in the range of about 10 mg to about 40 mg, or about 25 mg, of drug per dose subcutaneously biw for the duration of pirfenidone therapy; (2) a weight-based dosage of REMICADE® in the range of about 3 mg to about 10 mg of drug per kilogram of body weight intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks for the duration of pirfenidone therapy; or (3) a fixed dosage of HUMIRA™ of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks for the duration of pirfenidone therapy. Pirfenidone monotherapy for the treatment of ALD
[00226] In another aspect, the present invention provides pirfenidone or pirfenidone analog monotherapy therapy for the treatment of ALD.
[00227] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd, optionally in two or more divided doses per day, for the desired treatment duration.
[00228] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00229] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00230] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00231] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00232] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00233] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00234] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00235] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration.
[00236] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of ALD in a patient comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration. Pirfenidone combination therapy (other than IFN-γ) for the treatment of ALD
[00237] In another aspect, the present invention provides combination therapy for the treatment of ALD, comprising administering pirfenidone or a pirfenidone and at least a second therapeutic agent other than IFN-γ in combined amounts effective to treat ALD.
[00238] In some embodiments, the present invention provides a method using an effective amount of pirfenidone or a specific pirfenidone analog in the treatment of alcoholic hepatitis in a patient, comprising administering to the patient pirfenidone or a pirfenidone analog in a weight-based dosage in the range from about 5 mg/kg/day to about 125 mg/kg/day, or a fixed dosage of about 400 mg to about 3600 mg per day, or about 800 mg to about 2400 mg per day, or about 1000 mg to about 1800 mg per day, or about 1200 mg to about 1600 mg per day, administered orally in a single dose or two or more divided doses orally qd for the desired treatment duration; and an effective amount of a TNF-α antagonist. In these embodiments, the effective amount of TNF-α antagonist can be selected from: (1) a fixed dosage of ENBREL® in the range of about 10 mg to about 40 mg, or about 25 mg, of drug per dose subcutaneously biw for the duration of pirfenidone therapy; (2) a weight-based dosage of REMICADE® in the range of about 3 mg to about 10 mg of drug per kilogram of body weight intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks for the duration of pirfenidone therapy; or (3) a fixed dosage of HUMIRA™ of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks for the duration of pirfenidone therapy. Pirfenidone or a pirfenidone analog, a Type II interferon receptor agonist, and a side effect management agent in combination therapy to treat ALD
[00239] In one aspect, the present invention provides combination therapy for the treatment of ALD, comprising administering an effective amount of a Type II interferon receptor agonist, an effective amount of pirfenidone or a pirfenidone analog, and a side effect management agent. In some embodiments, the method employs a Type II interferon receptor agonist that is IFN-gamma (IFN-γ).
[00240] In one embodiment, the invention provides a method using an effective amount of IFN- γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising co-administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a dosage of pirfenidone or a pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight, or a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent, for the desired treatment duration.
[00241] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a dosage of pirfenidone or a pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00242] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00243] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 1200 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00244] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 800 mg to about 2400 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00245] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 1200 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00246] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 100 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00247] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 100 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 1200 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00248] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 100 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 800 mg to about 2400 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration. [00249] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 100 μg of drug per -dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 1200 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00250] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 200 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00251] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 200 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 1200 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00252] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 200 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 800 mg to about 2400 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00253] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a v agent in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 200 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, or biw, or per day substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 1200 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00254] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a total weekly dosage of IFN-gamma containing an amount of about 100 μg to about 1,500 μg of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or administered substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00255] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a total weekly dosage of IFN-gamma containing an amount of about 100 μg to about 1,500 μg of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or administered substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 1200 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00256] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a total weekly dosage of IFN-gamma containing an amount of about 100 μg to about 1,500 μg of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or administered substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 800 mg to about 2400 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00257] In another embodiment, the invention provides a method using an effective amount of IFN-γ, pirfenidone or a specific pirfenidone analog, and a side effect management agent in the treatment of ALD in a patient comprising administering to the patient a total weekly dosage of IFN-gamma containing an amount of about 100 μg to about 1,500 μg of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or administered substantially continuously or continuously; a fixed dosage of pirfenidone or a pirfenidone analog in the range of from about 400 mg to about 1200 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration; and a suitable dosage of a side effect management agent for the desired treatment duration.
[00258] In any of the above-described regimens, a side effect management agent can be selected from the group of aNSAID (e.g., aspirin, ibuprofen, acetaminophen); a histamine type 2 receptor antagonist; a hematopoietic agent; and an antacid. Suitable dosages for the use of such side effect management agents are well known in the art. For example, the dosages indicated on the label and/or package insert of any commercially available product having as its active ingredient any such side effect management agent can be used when practicing any of the above-described regimens. Type I interferon receptor agonist, Type II interferon receptor agonist, and TNF-α antagonist combination therapy in the treatment of ALD
[00259] In one aspect, the present invention provides combination therapy for the treatment of ALD, comprising administering an effective amount of a Type I interferon receptor agonist, an effective amount of a Type II interferon receptor agonist, and an effective amount of a TNF-α antagonist. In some embodiments, the method employs a Type II interferon receptor agonist that is IFN-gamma. In some embodiments, the method employs a Type I interferon receptor agonist that is IFN-α. In some embodiments, the methods further comprise administering an effective amount of pirfenidone or a pirfenidone analog.
[00260] In some embodiments, the invention provides methods using a synergistically effective amount of a Type I interferon receptor agonist, a Type II interferon receptor agonist, and a TNF-α antagonist in the treatment of ALD in a patient. In some embodiments, the invention provides methods using a synergistically effective amount of an IFN-α, IFN-γ, and a TNF-α antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRA™, in the treatment of ALD in a patient. In one embodiment, the invention provides a method using a synergistically effective amount of a consensus IFN-α, IFN-γ, and a TNF-α antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRA™, in the treatment of ALD in a patient.
[00261] In general, an effective amount of a consensus interferon (CIFN) and IFN-γ suitable for use in the methods of the invention is provided by a dosage ratio of 1 μg CIFN : 10 μg IFN-γ, where both CIFN and IFN-γ are unPEGylated and unglycosylated species.
[00262] In one embodiment, the invention provides a method using an effective amount of INFERGEN® consensus IFN-α, IFΝ-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of INFERGEN® containing an amount of about 1 μg to about 30 μg, of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN-γ containing an amount of about 10 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00263] In another embodiment, the invention provides a method using an effective amount of INFERGEN®consensus IFN-α, IFΝ-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of INFERGEN® containing an amount of about 1 μg to about 9 μg, of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN-γ containing an amount of about 10 μg to about 100 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once montiily, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration. [00264] In another embodiment, the invention provides a method using an effective amount of INFERGEN®consensus IFN-α, IFN-γ and TΝF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of INFERGEN® containing an amount of about 1 μg of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN-γ containing an amount of about 10 μg to about 50 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00265] In another embodiment, the invention provides a method using an effective amount of INFERGEN®consensus IFN-α, IFN-γ and TNF-α antagonist in the treatment of ALD in a patient comprising administering to the patient a dosage of INFERGEN® containing an amount of about 9 μg of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN-γ containing an amount of about 90 μg to about 100 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00266] In another embodiment, the invention provides a method using an effective amount of TΝFERGEN®consensus IFΝ-α, IFΝ-γ and TΝF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of INFERGEN® containing an amount of about 30 μg of drug per dose of INFERGEN®, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, a dosage of IFN-γ containing an amount of about 200 μg to about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once montiily, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00267] In another embodiment, the invention provides a method using an effective amount of PEGylated consensus IFN-α, IFN-γ and TΝF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEGylated consensus IFN-α (PEG-CIFN) containing an amount of about 4 μg to about 60 μg of CIFN amino acid weight per dose of PEG-CIFN, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN-γ containing an amount of about 30 μg to about 1,000 μg of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00268] In another embodiment, the invention provides a method using an effective amount of PEGylated consensus IFN-α, IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEGylated consensus IFN-α (PEG-CIFN) containing an amount of about 18 μg to about 24 μg of CIFN amino acid weight per dose of PEG-CIFN, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN-γ containing an amount of about 100 μg to about 300 μg of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration. [00269] In general, an effective amount of IFN-α 2a or 2b or 2c and IFN-γ suitable for use in the methods of the invention is provided by a dosage ratio of 1 million Units (MU) IFN-α 2a or 2b or 2c : 30 μg IFN-γ, where both IFN-α 2a or 2b or 2c and IFN-γ are unPEGylated and unglycosylated species.
[00270] In another embodiment, the invention provides a method using an effective amount of IFN-α 2a or 2b or 2c, IFN-γ and TΝF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFΝ-α 2a, 2b or 2c containing an amount of about 1 MU to about 20 MU of drug per dose of IFN-α 2a, 2b or 2c subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, a dosage of IFN-γ containing an amount of about 30 μg to about 600 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00271] In another embodiment, the invention provides a method using an effective amount of IFN-α 2a or 2b or 2c, IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN-α 2a, 2b or 2c containing an amount of about 3 MU of drug per dose of IFN-α 2a, 2b or 2c subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, a dosage of IFN-γ containing an amount of about 100 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug infravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00272] In another embodiment, the invention provides a method using an effective amount of IFN-α 2a or 2b or 2c, IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN-α 2a, 2b or 2c containing an amount of about 10 MU of drug per dose of IFN-α 2a, 2b or 2c subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, a dosage of IFN-γ containing an amount of about 300 μg of drug per dose of IFN-γ, subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once montiily, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00273] In another embodiment, the invention provides a method using an effective amount of PEGASYS®PEGylated IFN-α2a, IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEGASYS® containing an amount of about 90 μg to about 360 μg, of drug per dose of PEGASYS®, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN-γ containing an amount of about 30 μg to about 1,000 μg, of drug per week administered in divided doses subcutaneously qd, qod, tiw, or biw, or administered substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00274] In another embodiment, the invention provides a method using an effective amount of PEGASYS®PEGylated IFN-α2a, IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEGASYS® containing an amount of about 180 μg of drug per dose of PEGASYS®, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN-γ containing an amount of about 100 μg to about 300 μg, of drug per week administered in divided doses subcutaneously qd, qod, tiw, or biw, or administered substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00275] In another embodiment, the invention provides a method using an effective amount of PEG-LNTRON®PEGylated IFN-α2b, IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEG-INTRON® contaimng an amount of about 0.75 μg to about 3.0 μg of drug per kilogram of body weight per dose of PEG- INTRON®, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN-γ containing an amount of about 30 μg to about 1,000 μg of drug per week administered in divided doses subcutaneously qd, qod, tiw, or biw, or administered substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00276] In another embodiment, the invention provides a method using an effective amount of PEG-INTRON®PEGylated IFN-α2b, IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of PEG-INTRON® containing an amount of about 1.5 μg of drug per kilogram of body weight per dose of PEG-INTRON®, subcutaneously qw, qow, three times per month, or monthly, a total weekly dosage of IFN-γ containing an amount of about 100 μg to about 300 μg of drug per week administered in divided doses subcutaneously qd, qod, tiw, or biw, or administered substantially continuously or continuously, and a dosage of a TΝF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00277] In one embodiment, the present invention provides for a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 9 μg INFERGEN® consensus IFN-α administered subcutaneously qd or tiw; 50 μg Actimmune® hximan IFN-γ lb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow, for the desired treatment duration.
[00278] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 9 μg INFERGEN® consensus IFN-α administered subcutaneously qd or tiw; 100 μg Actimmune® human IFN-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/lcg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow, for the desired treatment duration.
[00279] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 9 μg INFERGEN® consensus IFN-α administered subcutaneously qd or tiw; 25 μg Actimmune® human IFN-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy.
[00280] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 9 μg INFERGEN® consensus IFN-α administered subcutaneously qd or tiw; 200 μg Actimmune® human IFN-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy.
[00281] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 100 μg monoPEG(30 kD, linear)- ylated consensus IFN-α administered subcutaneously every 10 days or qw; 50 μg Actimmune® human IFN-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy.
[00282] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to a patient a regimen of 100 μg monoPEG(30 kD, linear)- ylated consensus IFN-α administered subcutaneously every 10 days or qw; 100 μg Actimmune® human IFN-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy. .
[00283] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 150 μg monoPEG(30 kD, linear)- ylated consensus IFN-α administered subcutaneously every 10 days or qw; 50 μg Actimmune® human IFN-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy.
[00284] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 150 μg monoPEG(30 kD, linear)- ylated consensus IFN-α administered subcutaneously every 10 days or qw; 100 μg Actimmune® human IFN-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy.
[00285] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 200 μg monoPEG(30 kD, linear)- ylated consensus IFN-α administered subcutaneously every 10 days or qw; 50 μg Actimmune® human IFN-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy. [00286] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 200 μg monoPEG(30 kD, linear)- ylated consensus IFN-α administered subcutaneously every 10 days or qw; 100 μg Actimmune® human IFΝ-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered infravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy.
[00287] The therapeutic effect of the above regimens can be enhanced by co-administering to the patient a weight-based dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight, or a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg, or about 800 mg to about 2400 mg, or about 1000 mg to about 1800 mg, or about 1200 mg to about 1600 mg, orally qd for the desired duration of Type I interferon receptor agonist, Type II interferon receptor agonist and TΝF-α antagonist therapy. Type II interferon receptor agonist and TNF-α antagonist combination therapy in the treatment of ALD
[00288] In one aspect, the present invention provides combination therapy for the treatment of ALD, comprising administering an effective amount of a Type II interferon receptor agonist, and an effective amount of a TNF-α antagonist. In some embodiments, the method employs a Type II interferon receptor agonist that is IFΝ-gamma. In some embodiments, the methods further comprise administering an effective amount of pirfenidone or a pirfenidone analog.
[00289] In some embodiments, the invention provides methods using a synergistically effective amount of a Type II interferon receptor agonist and TΝF-α antagonist in the treatment of ALD in a patient. In some embodiments, the invention provides methods using a synergistically effective amount of IFΝ-γ and a TΝF-α antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRA™, in the treatment of ALD in a patient.
[00290] In one embodiment, the invention provides a method using an effective amount of IFN- γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 300 μg of drug per dose of IFN-γ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00291] In another embodiment, the invention provides a method using an effective amount of IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 100 μg of drug per dose of IFN-γ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00292] In another embodiment, the invention provides a method using an effective amount of IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN-γ containing an amount of about 10 μg to about 50 μg of drug per dose of IFN-γ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00293] In another embodiment, the invention provides a method using an effective amount of IFN-γ and TNF-α antagonist in the treatment of ALD in a patient comprising administering to the patient a dosage of IFN-γ containing an amount of about 90 μg to about 100 μg of drug per dose of IFN-γ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00294] In another embodiment, the invention provides a method using an effective amount of IFN-γ and TNF-α antagonist in the freatment of ALD in a patient, comprising administering to the patient a dosage of IFN-γ containing an amount of about 200 μg to about 300 μg of drug per dose of IFN-γ subcutaneously qd, qod, tiw, biw, qw, qow, three times per month, once monthly, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00295] In another embodiment, the invention provides a method using an effective amount of IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a total weekly dosage of IFN-γ containing an amount of about 30 μg to about 1,000 μg of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or substantially continuously or continuously, and a dosage of a TΝF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00296] In another embodiment, the invention provides a method using an effective amount of IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a total weekly dosage of IFN-γ containing an amount of about 100 μg to about 300 μg of drug per week in divided doses administered subcutaneously qd, qod, tiw, biw, or substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00297] In another embodiment, the invention provides a method using an effective amount of IFN-γ and TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of IFN-γ containing an amount of about 30 μg to about 600 μg of drug per dose of IFN-γ subcutaneously qd, qod, tiw, biw, or per day substantially continuously or continuously, and a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00298] The therapeutic effect or other therapeutic benefit of such regimens can be enhanced by co-administering to the patient a weight-based dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight, or a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg, or about 800 mg to about 2400 mg, or about 1000 mg to about 1800 mg, or about 1200 mg to about 1600 mg, orally qd for the desired duration of Type II interferon receptor agonist and TNF-α antagonist therapy.
[00299] In many embodiments, a Type II interferon receptor agonist and/or TNF-α antagonist is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time. In embodiments utilizing co-administration of pirfenidone or a specific pirfenidone analog, the duration of therapy with pirfenidone or a specific pirfenidone analog can be coincident with the duration of therapy with Type II interferon receptor agonist and/or TNF-α antagonist.
[00300] In one embodiment, the present invention provides for a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 50 μg Actimmune® human IFN-γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow, for the desired treatment duration.
[00301] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 100 μg Actimmune® human IFN- γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow, for the desired treatment duration.
[00302] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 25 μg Actimmune® human IFN- γlb administered subcutaneously tiw; and a dosage of TΝF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy.
[00303] In one embodiment, the present invention provides a method of treatment of ALD in a patient, comprising administering to the patient a regimen of 200 μg Actimmune® human IFN- γlb administered subcutaneously tiw; and a dosage of TNF-α antagonist selected from the group consisting of (i) 25 mg ENBREL® administered subcutaneously biw (ii) 3 mg REMICADE®/kg patient body weight administered intravenously at weeks 0, 2 and 6, and every 8 weeks thereafter or (iii) 40 mg HUMIRA™ subcutaneously qw or qow; for the desired duration of therapy. TΝF-α antagonist and pirfenidone or pirfenidone analog in combination therapy for the treatment of ALD
[00304] In one aspect, the present invention provides combination therapy for the treatment of ALD, comprising administering an effective amount of a TNF-α antagonist and an effective amount of pirfenidone or a pirfenidone analog.
[00305] In some embodiments, the invention provides methods using a synergistically effective amount of pirfenidone or a pirfenidone analog, and TNF-α antagonist in the treatment of ALD in a patient. In some embodiments, the invention provides methods using a synergistically effective amount of pirfenidone or a pirfenidone analog, and a TΝF-α antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRA™, in the treatment of ALD in a patient. In one embodiment, the invention provides a method using a synergistically effective amount of pirfenidone, and a TNF-α antagonist selected from the group consisting of ENBREL®, REMICADE® and HUMIRA™, in the treatment of ALD in a patient.
[00306] In some embodiments, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of a TNF-α antagonist selected from the group consisting of (i) ENBREL® in an amount of about 25 mg of drug subcutaneously biw (ii) REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks or (iii) HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration; and a dosage of pirfenidone or a pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight, or a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses, administered orally qd for the desired treatment duration.
[00307] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
[00308] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00309] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TΝF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a dosage of pirfenidone or a specific pirfenidone analog in the range of about 5 mg/kg of body weight to about 125 mg/kg of body weight orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00310] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
[00311] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the freatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00312] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TΝF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00313] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TΝF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired "treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
[00314] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00315] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 400 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00316] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drag subcutaneously biw, for the desired treatment duration.
[00317] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising admimstering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00318] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 800 mg to about 2400 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00319] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TΝF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
[00320] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00321] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00322] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
[00323] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00324] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1000 mg to about 1800 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00325] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising admimstering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
[00326] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TΝF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00327] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00328] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drug subcutaneously biw, for the desired treatment duration.
[00329] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drag intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00330] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfemdone analog in the range of about 1200 mg to about 1600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00331] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 1200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drag subcutaneously biw, for the desired treatment duration.
[00332] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of aboutl200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drug intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00333] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in an amount of about 1200 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00334] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1800 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of ENBREL® in an amount of about 25 mg of drag subcutaneously biw, for the desired treatment duration.
[00335] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1800 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of REMICADE® in an amount of about 3 mg/kg to about 10 mg/kg of drag intravenously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00336] In one embodiment, the invention provides a method using an effective amount of pirfenidone or a pirfenidone analog and a TNF-α antagonist in the treatment of ALD in a patient, comprising administering to the patient a fixed dosage of pirfenidone or a specific pirfenidone analog in the range of about 1800 mg to about 3600 mg in a single dose or two or three divided doses orally qd for the desired treatment duration; and a dosage of HUMIRA™ in an amount of about 40 mg of drug subcutaneously qw, qow, three times per month, once monthly, once every 6 weeks, or once every 8 weeks, for the desired treatment duration.
[00337] In many embodiments, pirfenidone or a pirfenidone analog and/or a TNF-α antagonist is administered for a period of about 1 day to about 7 days, or about 1 week to about 2 weeks, or about 2 weeks to about 3 weeks, or about 3 weeks to about 4 weeks, or about 1 month to about 2 months, or about 3 months to about 4 months, or about 4 months to about 6 months, or about 6 months to about 8 months, or about 8 months to about 12 months, or at least one year, and may be administered over longer periods of time. In embodiments utilizing co- administration of pirfenidone or a specific pirfenidone analog, the duration of therapy with pirfenidone or a specific pirfenidone analog can be coincident with the duration of therapy with TNF-α antagonist.
[00338] It will be understood that the above-described methods of treatment of ALD are suitable for the treatment of any stage of ALD in a patient. For example, the subject methods can be employed in the treatment of hepatic steatosis, alcoholic hepatitis, hepatic fibrosis, or hepatic cirrhosis, or any combination thereof, that occurs in a patient suffering from ALD.
[00339] In another example, the above-described methods of treatment of ALD can be employed in the treatment of non-fibrotic alcoholic hepatitis in a patient.

Claims

CLAIMSWhat is claimed is:
1. A method of treating non-fibrotic alcoholic hepatitis in an individual, the method comprising administering to the individual an effective amount of pirfenidone or a pirfenidone analog to ameliorate the disease.
2. The method of claim 1 , further comprising administering an effective amount of a Type If interferon receptor agonist.
3. The method of claim 2, further comprising administering an effective amount of a Type I interferon receptor agonist.
4. The method of any of claims 1 -3, further comprising administering an effective amount of a TNF-α antagonist other than pirfenidone or a pirfenidone analog.
5. A method of treating alcoholic liver disease in an individual, the method comprising administering to the individual an effective amount of pirfenidone or a pirfenidone analog to ameliorate the disease.
6. The method of claim 5, wherein the individual receives no IFN-γ during treatment with pirfenidone or pirfenidone analog.
7. A method of treating alcoholic liver disease in an individual, the method comprising administering to the individual an effective amount of a Type II interferon receptor agonist and pirfenidone or a pirfenidone analog to ameliorate the disease, and co- administering to the individual an amount of a third agent effective to reduce the incidence or severity of any side effect that would have been experienced by the individual in response to the administration of Type II interferon receptor agonist and pirfenidone or pirfenidone analog without co-administration of the third agent.
8. A method of treating alcoholic liver disease in a individual, the method comprising administering to the individual an effective amount of a TNF-α antagonist other than pirfenidone or a pirfenidone analog, and an effective amount of a Type II interferon receptor agonist, to ameliorate the disease.
9. The method of claim 8, further comprising administering pirfenidone or a pirfenidone analog.
10. The method of claim 8, further comprising administering an effective amount of a Type I interferon receptor antagonist.
11. A method of treating alcoholic liver disease in an individual, the method comprising administering to the individual an effective amount of a TNF-α antagonist other than pirfenidone or a pirfenidone analog and an effective amount of pirfenidone or a pirfenidone analog.
12. The method of any of claims 2, 7 and 8, wherein the Type II interferon receptor agonist is IFN-γ.
13. The method of claim 3 or 10, wherein the Type I interferon receptor agonist is an IFN-α.
14. The method of claim 13, wherein the IFN-α is a consensus interferon-α (CIFN).
15. The method of claim 13, wherein the IFN-α is a PEGylated IFN-α.
16. The method of claim 14, wherein the CIFN is a monoPEG (30 kD, linear)- ylated CIFN.
17. The method of any of claims 4, 8 and 11 , wherein the TNF-α antagonist is selected from etanercept, infliximab and adalimumab.
18. The method of any of claims 1-17, wherein the individual is a human.
PCT/US2004/035139 2003-10-24 2004-10-21 Method of treating alcoholic liver disease WO2005039598A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US51443803P 2003-10-24 2003-10-24
US60/514,438 2003-10-24

Publications (1)

Publication Number Publication Date
WO2005039598A1 true WO2005039598A1 (en) 2005-05-06

Family

ID=34520208

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/035139 WO2005039598A1 (en) 2003-10-24 2004-10-21 Method of treating alcoholic liver disease

Country Status (1)

Country Link
WO (1) WO2005039598A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008029145A1 (en) * 2006-09-07 2008-03-13 Ucl Business Plc Biqmarkers for assessing liver function
WO2010065755A1 (en) 2008-12-04 2010-06-10 Concert Pharmaceuticals, Inc. Deuterated pyridinones
US8969347B2 (en) 2008-06-03 2015-03-03 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
US9359379B2 (en) 2012-10-02 2016-06-07 Intermune, Inc. Anti-fibrotic pyridinones
US9527816B2 (en) 2005-05-10 2016-12-27 Intermune, Inc. Method of modulating stress-activated protein kinase system
US10188637B2 (en) 2016-03-29 2019-01-29 Hoffmann-La Roche Inc. Granulate formulation of 5-methyl-1-phenyl-2-(1H)-pyridone and method of making the same
US10233195B2 (en) 2014-04-02 2019-03-19 Intermune, Inc. Anti-fibrotic pyridinones

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5952309A (en) * 1995-09-29 1999-09-14 Eisai Company, Ltd. Method for treating alcoholic liver disease

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5952309A (en) * 1995-09-29 1999-09-14 Eisai Company, Ltd. Method for treating alcoholic liver disease

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9527816B2 (en) 2005-05-10 2016-12-27 Intermune, Inc. Method of modulating stress-activated protein kinase system
US10010536B2 (en) 2005-05-10 2018-07-03 Intermune, Inc. Method of modulating stress-activated protein kinase system
US8455220B2 (en) 2006-09-07 2013-06-04 Ucl Business Plc Biomarkers for assessing liver function
WO2008029145A1 (en) * 2006-09-07 2008-03-13 Ucl Business Plc Biqmarkers for assessing liver function
US8969347B2 (en) 2008-06-03 2015-03-03 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
US9290450B2 (en) 2008-06-03 2016-03-22 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
USRE47142E1 (en) 2008-06-03 2018-11-27 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
WO2010065755A1 (en) 2008-12-04 2010-06-10 Concert Pharmaceuticals, Inc. Deuterated pyridinones
US9359379B2 (en) 2012-10-02 2016-06-07 Intermune, Inc. Anti-fibrotic pyridinones
US9675593B2 (en) 2012-10-02 2017-06-13 Intermune, Inc. Anti-fibrotic pyridinones
US10376497B2 (en) 2012-10-02 2019-08-13 Intermune, Inc. Anti-fibrotic pyridinones
US10898474B2 (en) 2012-10-02 2021-01-26 Intermune, Inc. Anti-fibrotic pyridinones
US10233195B2 (en) 2014-04-02 2019-03-19 Intermune, Inc. Anti-fibrotic pyridinones
US10544161B2 (en) 2014-04-02 2020-01-28 Intermune, Inc. Anti-fibrotic pyridinones
US10188637B2 (en) 2016-03-29 2019-01-29 Hoffmann-La Roche Inc. Granulate formulation of 5-methyl-1-phenyl-2-(1H)-pyridone and method of making the same

Similar Documents

Publication Publication Date Title
US20090068142A1 (en) Compositions and methods for treating coronavirus infection and sars
WO2006016930A2 (en) Methods for treating hcv infection
US20090226400A1 (en) Continuous delivery methods for treating hepatitis virus infection
US20070072181A1 (en) Combination therapy for treating alphavirus infection and liver fibrosis
CA2460690A1 (en) Method of treating hepatitis virus infection with a multiphasic interferon delivery profile
US20050095224A1 (en) Compositions and method for treating hepatitis virus infection
US20070258946A1 (en) Combination Therapy for Treating Hepatitis C Virus Infection
WO2004078194A1 (en) Interferon drug therapy for the treatment of viral diseases and liver fibrosis
WO2005038056A1 (en) Combination therapy for the treatment of viral diseases
US20070032457A1 (en) Combination therapy for cancer treatment
WO2005110455A2 (en) Combination therapy for treating hepatitis virus infection
WO2005110478A2 (en) Combination therapy for treating fibrotic disorders
WO2005039598A1 (en) Method of treating alcoholic liver disease
WO2005062949A2 (en) Method for treating hepatitis virus infection
WO2004078207A1 (en) Interferon drug therapy for the treatment of viral diseases and liver fibrosis
US20060198823A1 (en) Compositions and methods for treating viral infections
US20090035273A1 (en) Combination treatment method with interferon-tau
EP1251866A1 (en) Combination of temozolomide and pegylated interferon-alpha for treating cancer
US20070154454A1 (en) Compositions and methods for treating poxvirus infection
MXPA04003239A (en) Methods of treating liver fibrosis and hepatitis c virus infection.
US20070231301A1 (en) Parenteral low dose type 1 interferons for bladder cancer
MXPA00003346A (en) Chronic myeloid leukemia (cml) therapy

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase