WO2003045430A1 - Tnf-alpha inhibitors for the treatment of hepatic diseases - Google Patents

Tnf-alpha inhibitors for the treatment of hepatic diseases Download PDF

Info

Publication number
WO2003045430A1
WO2003045430A1 PCT/GB2002/005300 GB0205300W WO03045430A1 WO 2003045430 A1 WO2003045430 A1 WO 2003045430A1 GB 0205300 W GB0205300 W GB 0205300W WO 03045430 A1 WO03045430 A1 WO 03045430A1
Authority
WO
WIPO (PCT)
Prior art keywords
tnfα
inhibitor
hepatic disease
treatment
use according
Prior art date
Application number
PCT/GB2002/005300
Other languages
French (fr)
Inventor
Gabriel Stavros Panayi
Original Assignee
King's College London
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 King's College London filed Critical King's College London
Priority to AU2002343085A priority Critical patent/AU2002343085A1/en
Publication of WO2003045430A1 publication Critical patent/WO2003045430A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • 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
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to the use of TNF ⁇ inhibitors in the treatment of hepatic diseases.
  • Hepatic diseases include any inflammatory disease of the liver. In humans the most common cause is viral infection of the liver by viruses such as hepatitis A, B and C, although there are other viruses that cause hepatitis, including yellow fever virus, Epstein-Barr virus and cytomegalo virus. Hepatic diseases cause liver cell damage, necrosis and eventual liver fibrosis or the development of one or more liver hepatomas which may be lethal.
  • Hepatic diseases can also be caused by amoeba infections, alcohol and substance abuse.
  • Amoeba infections can also be caused by amoeba infections, alcohol and substance abuse.
  • Chitturi et al. Semin. Liver Dis., 22, 169-183, 2002.
  • the pathogenesis of the hepatic diseases is a combination of the effects of the causative agent, e.g. the virus, and of the host immune and inflammatory response.
  • the treatment of hepatic diseases, especially viral hepatitis, is difficult, expensive and time consuming. Furthermore, treatments are not always successful.
  • Cytokines are involved in the host's immune response by inducing apoptosis of liver cells but, paradoxically, at the same time inhibiting virus production (Kaplowitz et al., Semin. Liver Dis., 22, 137-144, 2002; Jaeschke et al, Toxicol. ScL, 65, 166-176, 2002; Neuman et al, Alcohol Clin. Exp. Res., 25 (S Suppl ISBRA) 251S-253S, 2001; and Neuman, Crit. Rev. Clin Lab. Sci., 38, 109-166, 2001).
  • TNF ⁇ cytokine tumour necrosis factor ⁇
  • the present invention relates to the use of anti-TNF ⁇ therapy for the treatment of hepatitis D, C, and G, and all other types and forms of viral hepatitis, and alcoholic hepatitis.
  • the present invention provides the use of a TNF ⁇ inhibitor in the treatment of a hepatic disease.
  • the present invention also provides the use of a TNF ⁇ inhibitor in the manufacture of a medicament for the treatment of a hepatic disease.
  • the present invention also provide a method of treating a hepatic disease comprising administrating to a patient in need thereof an effective amount of a TNF ⁇ inhibitor.
  • TNF ⁇ inhibitor refers to any agent that inhibits the inflammatory action of TNF ⁇ .
  • the TNF ⁇ inhibitor may inhibit the production, secretion or activity of TNF ⁇ thereby preventing the inflammatory action.
  • the agent may be a protein, a nucleic acid, a low molecular weight organic or inorganic compound, or a carbohydrate.
  • the agent may directly interact with TNF ⁇ or may inhibit the inflammatory action by blocking the relevant TNF ⁇ receptor or may inhibit the inflammatory action further down the inflammatory pathway.
  • the TNF ⁇ inhibitor may inhibit the production of TNF ⁇ by preventing its expression or secretion.
  • the TNF ⁇ inhibitor may also target TNF ⁇ for degradation reducing the level of active TNF ⁇ in the liver.
  • the TNF ⁇ inhibitor may cause genetic inhibition of TNF ⁇ secretion or action by gene therapy or by interfering with the TNF ⁇ encoding RNA.
  • the TNF ⁇ inhibitor interacts with TNF ⁇ and prevents or reduces its ability to cause inflammation.
  • the TNF ⁇ inhibitor inhibits TNF ⁇ binding to its receptor.
  • the TNF ⁇ inhibitor may do this by interacting with TNF ⁇ or with the receptor.
  • Suitable TNF ⁇ inhibitors include antibody molecules or soluble TNF ⁇ receptors that bind to TNF ⁇ .
  • the soluble TNF ⁇ receptor is a soluble p75 TNF-receptor.
  • the TNF ⁇ inhibitor may be coupled to a moiety that increase the half life in the body and so increases the effectiveness of the inhibitor, decreases the quantity administered and decreases the frequency of administration.
  • suitable moieties include Fc fragments of human immunoglobulins, especially a IgG immunoglobulin Fc fragment, polyethylene glycol (PEG) molecules, etc.
  • the TNF ⁇ inhibitor is an antibody molecule.
  • the antibody molecule may be any antibody molecule capable of specifically binding to TNF ⁇ .
  • the antibody molecule may be a complete polyclonal or monoclonal antibody molecule or antigen binding fragment thereof, such as Fv, Fab, F(ab') 2 fragments and single chain Fv fragments thereof.
  • the antibody molecule may be a recombinant antibody molecule such as a chimeric antibody molecule, preferably having human constant regions and mouse variable regions, or a CDR grafted antibody molecule, or antigen binding fragment thereof. Methods for producing such antibody molecules are well known to those skilled in the art and are described in EP-A-0120694 and EP-A-0125023.
  • the anti-TNF ⁇ therapy may consist of purified antibodies of human or other animal source, genetically engineered complete antibodies or chimeric antibodies or partial forms of antibodies, or constructs of TNF ⁇ soluble receptors with human Fc or chemically modified and small molecular weight compounds that inhibit production or secretion or action of TNF ⁇ .
  • the TNF ⁇ inhibitor is the chimeric (mouse/human) antibody against tumour necrosis factor alpha (TNF ⁇ ) InfliximabTM (Schering-Plough).
  • TNF ⁇ tumour necrosis factor alpha
  • a hepatic disease refers to any hepatic disease, i.e. any disease that causes inflammatory damage to the liver, wherein the damage is accentuated by TNF ⁇ .
  • the hepatic disease may be caused by a viral agent, an amoeba or other parasite, alcoholism, cancers, drug overdoses, narcotic abuse,
  • the hepatic disease is caused by a viral agent.
  • the viral agent is a hepatitis A, B, C, D, E, F or G; Epstein-Barr virus, cytomegalovirus, herpes virus, or the yellow fever virus. It is particularly preferred that the viral agent is hepatitis B, D, C or G, most preferably hepatitis B.
  • the TNF ⁇ inhibitor may be formulated in combination with any pharmaceutically acceptable carrier, adjuvant or vehicle.
  • Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat.
  • the TNF ⁇ inhibitor may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. Preferably the TNF ⁇ inhibitor is administered orally or by injection.
  • the TNF ⁇ inhibitor may be formulated with any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles.
  • parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
  • the TNF ⁇ inhibitor may be administered in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension.
  • This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • suitable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • oils such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as Ph. Helv or a similar alcohol.
  • the TNF ⁇ inhibitor may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions.
  • carriers which are commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried corn starch.
  • the TNF ⁇ inhibitor may also be administered in the form of suppositories for rectal administration.
  • These compositions can be prepared by mixing a compound of this invention with a suitable non- irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components.
  • suitable non- irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • the TNF ⁇ inhibitor may be administered by nasal aerosol or inhalation.
  • compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents l ⁇ iown in the art.
  • TNF ⁇ inhibitor The appropriate dosage of the TNF ⁇ inhibitor will vary depending upon, for example, the nature and severity of the disorder to be treated and the mode of administration. For example, a dosage of about lmg/kg to about lOmg/kg body weight given every 2 to 4 weeks will satisfactorily treat a hepatic disease.
  • Figure 1 shows the reduction in hepatitis B DNA levels during treatment with a TNF ⁇ inhibitor.
  • H. pylori positive gastritis Treatment with non-steroidal anti-inflammatory drugs was limited by H. pylori positive gastritis. Intra-muscular injections of methyl prednisolone were initiated. Their benefit was limited. Intra-muscular gold injections were commenced in October 1994 as a result of generally worsening disease activity. He was admitted with severe hip and shoulder disease with an associated dactylitis. The ESR was 88 with an alkaline phosphatase of 138, ALT 35 ( ⁇ 34) and gamma glutamine transferase ( ⁇ GT) 104 ( ⁇ 50). Hepatitis serology gave a positive surface antigen and antibodies to HepBe but no HepBe or Core IgM. This suggested hepatitis B with low infectivity.
  • the dose infused was 3 mg/kg.
  • All measures of disease activity improved (Health Assessment Questionnaire, ESR, CRP, early morning stiffness, global assessment, tender and swollen joint counts and the Ritchie Index). He became pain free. His psoriatic skin disease settled. The liver function tests also returned to within the no ⁇ nal limits. A decision was made to monitor the Hepatitis B DNA levels during treatment. Hepatitis B DNA, measured after the fourth infusion of InfliximabTM, was undetectable.
  • TNF ⁇ may be achieved in a variety of ways.
  • Any agent that is capable of inhibiting the binding of TNF ⁇ to its receptor may be used.
  • agent may be an antibody which is capable of binding to TNF ⁇ or fragment of such antibody or it may be a soluble p75 TNF-receptor or fragment thereof.
  • These agents are capable of binding to TNF ⁇ and inhibiting TNF ⁇ binding to the TNF receptors present on cells.
  • agents may also be coupled to other protein moieties to increase their half life in the body and so increase their effectiveness, by decreasing the quantity administered or the frequency of administration of the reagent to the patient.
  • protein moieties which may be used to have this effect are the Fc fragment of human immunoglobulins especially where such Fc fragment is derived from human immuno globulin and more preferably where the Fc fragment is derived from a human IgG class immunoglobulin.
  • the half life of the agent to be used may also be increased by treating the preparation with polyethylene glycol.

Abstract

The present invention relates to the use of TNFα inhibitors in the treatment of viral hepatic diseases.

Description

TNF-ALPHA INHIBITORS FOR THE TREATMENT OF HEPATIC DISEASES
The present invention relates to the use of TNFα inhibitors in the treatment of hepatic diseases.
Hepatic diseases include any inflammatory disease of the liver. In humans the most common cause is viral infection of the liver by viruses such as hepatitis A, B and C, although there are other viruses that cause hepatitis, including yellow fever virus, Epstein-Barr virus and cytomegalo virus. Hepatic diseases cause liver cell damage, necrosis and eventual liver fibrosis or the development of one or more liver hepatomas which may be lethal.
Hepatic diseases can also be caused by amoeba infections, alcohol and substance abuse. For a review see Chitturi et al., Semin. Liver Dis., 22, 169-183, 2002.
The complications of viral hepatitis therefore are many and affect millions of people throughout the world.
The pathogenesis of the hepatic diseases is a combination of the effects of the causative agent, e.g. the virus, and of the host immune and inflammatory response. The treatment of hepatic diseases, especially viral hepatitis, is difficult, expensive and time consuming. Furthermore, treatments are not always successful.
Cytokines are involved in the host's immune response by inducing apoptosis of liver cells but, paradoxically, at the same time inhibiting virus production (Kaplowitz et al., Semin. Liver Dis., 22, 137-144, 2002; Jaeschke et al, Toxicol. ScL, 65, 166-176, 2002; Neuman et al, Alcohol Clin. Exp. Res., 25 (S Suppl ISBRA) 251S-253S, 2001; and Neuman, Crit. Rev. Clin Lab. Sci., 38, 109-166, 2001). However, there is also evidence that treating patients with chronic hepatitis C with interferon α leads to increased levels of the cytokine tumour necrosis factor α (TNFα) in the serum and this has been taken to mean that interferon α treatment may be accentuating inflammatory damage in the liver as evidenced by the release of TNFα.
It has now been found that by blocking TNFα the inflammatory element of hepatic diseases can be removed.
Accordingly, the present invention relates to the use of anti-TNFα therapy for the treatment of hepatitis D, C, and G, and all other types and forms of viral hepatitis, and alcoholic hepatitis.
In particular, the present invention provides the use of a TNFα inhibitor in the treatment of a hepatic disease.
The present invention also provides the use of a TNFα inhibitor in the manufacture of a medicament for the treatment of a hepatic disease.
The present invention also provide a method of treating a hepatic disease comprising administrating to a patient in need thereof an effective amount of a TNFα inhibitor.
The term "TNFα inhibitor" as used herein refers to any agent that inhibits the inflammatory action of TNFα. The TNFα inhibitor may inhibit the production, secretion or activity of TNFα thereby preventing the inflammatory action. The agent may be a protein, a nucleic acid, a low molecular weight organic or inorganic compound, or a carbohydrate. The agent may directly interact with TNFα or may inhibit the inflammatory action by blocking the relevant TNFα receptor or may inhibit the inflammatory action further down the inflammatory pathway. Furthermore, the TNFα inhibitor may inhibit the production of TNFα by preventing its expression or secretion. The TNFα inhibitor may also target TNFα for degradation reducing the level of active TNFα in the liver. The TNFα inhibitor may cause genetic inhibition of TNFα secretion or action by gene therapy or by interfering with the TNFα encoding RNA. Preferably the TNFα inhibitor interacts with TNFα and prevents or reduces its ability to cause inflammation. Preferably the TNFα inhibitor inhibits TNFα binding to its receptor. The TNFα inhibitor may do this by interacting with TNFα or with the receptor. Suitable TNFα inhibitors include antibody molecules or soluble TNFα receptors that bind to TNFα. Preferably the soluble TNFα receptor is a soluble p75 TNF-receptor.
The TNFα inhibitor may be coupled to a moiety that increase the half life in the body and so increases the effectiveness of the inhibitor, decreases the quantity administered and decreases the frequency of administration. Examples of suitable moieties include Fc fragments of human immunoglobulins, especially a IgG immunoglobulin Fc fragment, polyethylene glycol (PEG) molecules, etc.
It is most preferred that the TNFα inhibitor is an antibody molecule. The antibody molecule may be any antibody molecule capable of specifically binding to TNFα. The antibody molecule may be a complete polyclonal or monoclonal antibody molecule or antigen binding fragment thereof, such as Fv, Fab, F(ab')2 fragments and single chain Fv fragments thereof. The antibody molecule may be a recombinant antibody molecule such as a chimeric antibody molecule, preferably having human constant regions and mouse variable regions, or a CDR grafted antibody molecule, or antigen binding fragment thereof. Methods for producing such antibody molecules are well known to those skilled in the art and are described in EP-A-0120694 and EP-A-0125023.
As indicated above, the anti-TNFα therapy may consist of purified antibodies of human or other animal source, genetically engineered complete antibodies or chimeric antibodies or partial forms of antibodies, or constructs of TNFα soluble receptors with human Fc or chemically modified and small molecular weight compounds that inhibit production or secretion or action of TNFα.
In a particularly preferred embodiment of the present invention the TNFα inhibitor is the chimeric (mouse/human) antibody against tumour necrosis factor alpha (TNFα) Infliximab™ (Schering-Plough). The term "a hepatic disease" refers to any hepatic disease, i.e. any disease that causes inflammatory damage to the liver, wherein the damage is accentuated by TNFα. The hepatic disease may be caused by a viral agent, an amoeba or other parasite, alcoholism, cancers, drug overdoses, narcotic abuse,
Preferably the hepatic disease is caused by a viral agent. Preferably the viral agent is a hepatitis A, B, C, D, E, F or G; Epstein-Barr virus, cytomegalovirus, herpes virus, or the yellow fever virus. It is particularly preferred that the viral agent is hepatitis B, D, C or G, most preferably hepatitis B.
The TNFα inhibitor may be formulated in combination with any pharmaceutically acceptable carrier, adjuvant or vehicle. Pharmaceutically acceptable carriers, adjuvants and vehicles that may be used include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat.
The TNFα inhibitor may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. Preferably the TNFα inhibitor is administered orally or by injection. The TNFα inhibitor may be formulated with any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intra-articular, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques. The TNFα inhibitor may be administered in the form of a sterile injectable preparation, for example, as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant such as Ph. Helv or a similar alcohol.
The TNFα inhibitor may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, and aqueous suspensions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral admimstration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions are administered orally, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring and/or coloring agents may be added.
The TNFα inhibitor may also be administered in the form of suppositories for rectal administration. These compositions can be prepared by mixing a compound of this invention with a suitable non- irritating excipient which is solid at room temperature but liquid at the rectal temperature and therefore will melt in the rectum to release the active components. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols. The TNFα inhibitor may be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents lαiown in the art.
The appropriate dosage of the TNFα inhibitor will vary depending upon, for example, the nature and severity of the disorder to be treated and the mode of administration. For example, a dosage of about lmg/kg to about lOmg/kg body weight given every 2 to 4 weeks will satisfactorily treat a hepatic disease.
The present invention is now described by way of example only, with reference to the accompanying figure.
Figure 1 shows the reduction in hepatitis B DNA levels during treatment with a TNFα inhibitor.
EXAMPLE
We have successfully treated a 51 year old Vietnamese refugee with poorly controlled psoriatic arthritis (PsA). He presented to the Rheumatology Department at Guy's and St Thomas' Hospital with a long history of back pain and a raised ESR in 1993. Initial investigations included a normal FBC, ESR 53 (normal <15), alkaline phosphatase 137 (<115) and positive HLA B27. X-rays showed obliteration of the sacro-iliac joints with early syndesmophytes and lumbar squaring. A diagnosis of ankylosing spondylitis was made. In December 1993 a rash was noted. This had previously been present at the age of 10 years. The rash was suggestive of psoriasis and the diagnosis was confirmed at the St John's Dermatology Hospital.
Treatment with non-steroidal anti-inflammatory drugs was limited by H. pylori positive gastritis. Intra-muscular injections of methyl prednisolone were initiated. Their benefit was limited. Intra-muscular gold injections were commenced in October 1994 as a result of generally worsening disease activity. He was admitted with severe hip and shoulder disease with an associated dactylitis. The ESR was 88 with an alkaline phosphatase of 138, ALT 35 (<34) and gamma glutamine transferase (γGT) 104 (<50). Hepatitis serology gave a positive surface antigen and antibodies to HepBe but no HepBe or Core IgM. This suggested hepatitis B with low infectivity. Unfortunately the gold injections had to be stopped due to worsening of the skin rash. Liver biopsy performed in 1995 showed chronic active hepatitis. Blood hepatitis B DNA was positive at 66 pg/ml. Liver function tests deteriorated with alkaline phosphatase 224, total bilirubin 45 (<15), ALT 1522 and γGT 144. Over the next two years he took increasing doses of sulphasalazine and prednisolone. June 1998 saw the addition of Cyclosporin A (CsA) for deteriorating skin and joint disease. Hepatitis B serology showed Hep B surface antigen positive, core antibody positive, E antigen negative and E antibody positive. Liver biopsy repeated in September 1998 again showed chronic persistent hepatitis. The treatment escalated to include intravenous boluses of methyl-prednisolone, deflazacort 442 mg, CsA 75 mg and 50 mg per day and the introduction of methotrexate. CsA was stopped due to deteriorating renal function and hypertension. Treatment with methotrexate was suspended for a few months as LFTs deteriorated.
Despite methotrexate 12.5 mg/week and 30 mg deflazacort daily, he continued to have poorly controlled skin disease and joint signs and symptoms. He developed multiple complications secondary to treatment. These included corticosteroids induced osteoporosis, hypertension, mild renal impairment and the need for bilateral hip replacements. He also had chronic hepatitis B infection. He remained in significant pain and restricted in activities of daily living. Therapeutic options were limited. The use of leflunomide (Arava) was rejected as result of its potential for further liver disturbance. Despite the risk of re-awakening HBV, it was decided that Infliximab™ (Schering-Plough) should be administered.
He received 4 infusions of anti-TNFα therapy with Infliximab™, given as an intra-venous infusion on weeks 0, 2, 6 and then 8 weekly. The dose infused was 3 mg/kg. During treatment all measures of disease activity improved (Health Assessment Questionnaire, ESR, CRP, early morning stiffness, global assessment, tender and swollen joint counts and the Ritchie Index). He became pain free. His psoriatic skin disease settled. The liver function tests also returned to within the noπnal limits. A decision was made to monitor the Hepatitis B DNA levels during treatment. Hepatitis B DNA, measured after the fourth infusion of Infliximab™, was undetectable.
A summary of the patients status is set out in Table 1 below.
Table 1
Figure imgf000009_0001
One concern in this case was whether by removing the pro-inflammatory response Hep-B viral replication would become uncontrolled. This has not occurred. On the contrary, anti-TNFα appears to have stopped viral replication, as measured by blood Hep B DNA, for some weeks without further treatment.
It will be appreciated that inhibition of the action of TNFα may be achieved in a variety of ways. Any agent that is capable of inhibiting the binding of TNFα to its receptor may be used. Such agent may be an antibody which is capable of binding to TNFα or fragment of such antibody or it may be a soluble p75 TNF-receptor or fragment thereof. These agents are capable of binding to TNFα and inhibiting TNFα binding to the TNF receptors present on cells. Such agents may also be coupled to other protein moieties to increase their half life in the body and so increase their effectiveness, by decreasing the quantity administered or the frequency of administration of the reagent to the patient. Examples of protein moieties which may be used to have this effect are the Fc fragment of human immunoglobulins especially where such Fc fragment is derived from human immuno globulin and more preferably where the Fc fragment is derived from a human IgG class immunoglobulin. The half life of the agent to be used may also be increased by treating the preparation with polyethylene glycol.
All documents cited above are incorporated herein by reference.

Claims

Claims
1. Use of a TNFα inhibitor in the treatment of a hepatic disease.
2. Use of a TNFα inhibitor in the manufacture of a medicament for the treatment of a hepatic disease.
3. A method of treating a hepatic disease comprising administrating to a patient in need thereof an effective amount of a TNFα inhibitor.
4. The use according to claim 1 or claim 2, or the method according to claim 3, wherein the TNFα inhibitor inhibits the binding of TNFα to its receptor.
5. The use according to claim 1 or claim 2, or the method according to claim 3, wherein the TNFα inhibitor is an antibody molecule which binds to TNFα.
6. The use according to claim 1 or claim 2, or the method according to claim 3, wherein the TNFα inhibitor is Infliximab™.
7. The use according to claim 1 or claim 2, or the method according to claim 3, wherein the TNFα inhibitor is a soluble TNFα receptor.
8. The use according to claim 1 or claim 2, or the method according to claim 3, wherein the TNFα inhibitor comprises a moiety that increases the half life of the inhibitor in the body.
9. The use according to claim 1 or claim 2, or the method according to claim 3, wherein the hepatic disease hepatic disease is caused by a viral agent.
10. The use according to claim 1 or claim 2, or the method according to claim 3, wherein the hepatic disease is caused by hepatitis A, B, C, D, E, F or G; Epstein-Barr virus, cytomegalo virus, herpes virus, or the yellow fever virus.
11. The use accordmg to claim 1 or claim 2, or the method according to claim 3, wherein the hepatic disease is caused by hepatitis B, D, C or G.
12. The use according to claim 1 or claim 2, or the method according to claim 3, wherein the hepatic disease is caused by hepatitis B.
13. The use of an inhibitor of the action of Tumour necrosis factor (TNFα) for the preparation of a medicament for the treatment of viral hepatic disease.
14. The use according to claim 13 in which the inhibitor is one which inhibits the binding of TNFα to its receptor.
15. The use according to claim 13 in which the inhibitor is an antibody which binds to TNFα or a binding fragment of said antibody.
16. The use according to claim 13, in which the inhibitor is a soluble TNF -receptor which binds to TNFα or a binding fragment of said receptor.
17. A method of treatment of a viral hepatic disease which comprises administering to a patient in need thereof an inhibitor of the action of TNFα.
PCT/GB2002/005300 2001-11-23 2002-11-25 Tnf-alpha inhibitors for the treatment of hepatic diseases WO2003045430A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002343085A AU2002343085A1 (en) 2001-11-23 2002-11-25 Tnf-alpha inhibitors for the treatment of hepatic diseases

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0128138.5 2001-11-23
GBGB0128138.5A GB0128138D0 (en) 2001-11-23 2001-11-23 Pharmaceutical use

Publications (1)

Publication Number Publication Date
WO2003045430A1 true WO2003045430A1 (en) 2003-06-05

Family

ID=9926358

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2002/005300 WO2003045430A1 (en) 2001-11-23 2002-11-25 Tnf-alpha inhibitors for the treatment of hepatic diseases

Country Status (3)

Country Link
AU (1) AU2002343085A1 (en)
GB (1) GB0128138D0 (en)
WO (1) WO2003045430A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005117966A1 (en) * 2004-06-04 2005-12-15 Apoxis Sa Method to reduce hepatotoxicity of fas-mediated apoptosis-inducing agents
US9028822B2 (en) 2002-06-28 2015-05-12 Domantis Limited Antagonists against TNFR1 and methods of use therefor

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5753628A (en) * 1995-06-07 1998-05-19 Centocor, Inc. Peptide inhibitors of TNF containing predominantly D-amino acids
WO1998022137A1 (en) * 1996-11-15 1998-05-28 The Kennedy Institute Of Rheumatology SUPPRESSION OF TNF α AND IL-12 IN THERAPY
WO1999009965A2 (en) * 1997-08-21 1999-03-04 Takeda Chemical Industries, Ltd. Anti-inflammatory agent
JPH11127882A (en) * 1997-10-27 1999-05-18 Nippon Kayaku Co Ltd Novel physiologically active substance nk30424a and nk3024 b, their production and use
JPH11228576A (en) * 1997-12-10 1999-08-24 Japan Tobacco Inc Apoptosis inhibitor
WO1999064419A1 (en) * 1998-06-10 1999-12-16 Aventis Pharma S.A. Pyrrole derivatives, preparation method and pharmaceutical compositions containing same
US6107273A (en) * 1995-01-24 2000-08-22 Thomas Jefferson University Tumor necrosis factor inhibitors
EP1029851A1 (en) * 1997-10-14 2000-08-23 Yoshitomi Pharmaceutical Industries, Ltd. Piperazine compounds and medicinal use thereof
WO2000062790A2 (en) * 1999-04-19 2000-10-26 Immunex Corporation Soluble tumor necrosis factor receptor treatment of medical disorders
JP2001081088A (en) * 1999-09-10 2001-03-27 Nippon Kayaku Co Ltd New physiologically active substance nk3042 analogue and production and use thereof
US6284471B1 (en) * 1991-03-18 2001-09-04 New York University Medical Center Anti-TNFa antibodies and assays employing anti-TNFa antibodies

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6284471B1 (en) * 1991-03-18 2001-09-04 New York University Medical Center Anti-TNFa antibodies and assays employing anti-TNFa antibodies
US6107273A (en) * 1995-01-24 2000-08-22 Thomas Jefferson University Tumor necrosis factor inhibitors
US5753628A (en) * 1995-06-07 1998-05-19 Centocor, Inc. Peptide inhibitors of TNF containing predominantly D-amino acids
WO1998022137A1 (en) * 1996-11-15 1998-05-28 The Kennedy Institute Of Rheumatology SUPPRESSION OF TNF α AND IL-12 IN THERAPY
WO1999009965A2 (en) * 1997-08-21 1999-03-04 Takeda Chemical Industries, Ltd. Anti-inflammatory agent
EP1029851A1 (en) * 1997-10-14 2000-08-23 Yoshitomi Pharmaceutical Industries, Ltd. Piperazine compounds and medicinal use thereof
JPH11127882A (en) * 1997-10-27 1999-05-18 Nippon Kayaku Co Ltd Novel physiologically active substance nk30424a and nk3024 b, their production and use
JPH11228576A (en) * 1997-12-10 1999-08-24 Japan Tobacco Inc Apoptosis inhibitor
WO1999064419A1 (en) * 1998-06-10 1999-12-16 Aventis Pharma S.A. Pyrrole derivatives, preparation method and pharmaceutical compositions containing same
WO2000062790A2 (en) * 1999-04-19 2000-10-26 Immunex Corporation Soluble tumor necrosis factor receptor treatment of medical disorders
JP2001081088A (en) * 1999-09-10 2001-03-27 Nippon Kayaku Co Ltd New physiologically active substance nk3042 analogue and production and use thereof

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
CZAJA M J (REPRINT) ET AL: "TREATMENT WITH SOLUBLE TUMOR - NECROSIS - FACTOR - ALPHA RECEPTOR (STNFR) REDUCES TOXIC LIVER -INJURY", HEPATOLOGY, ( OCT 1993 ) VOL. 18, NO. 4, PART 2, PP. A111. ISSN: 0270-9139., YESHIVA UNIV ALBERT EINSTEIN COLL MED, MARION BESSIN LIVER RES CTR, BRONX, NY, 10461, XP008013047 *
DATABASE WPI Section Ch Week 199930, Derwent World Patents Index; Class B04, AN 1999-350332, XP002230418 *
DATABASE WPI Section Ch Week 200144, Derwent World Patents Index; Class B03, AN 2001-412369, XP002230419 *
MENON K V (REPRINT) ET AL: "Pilot study of the safety and tolerability of a monoclonal anti- tumor necrosis factor alpha antibody in patients with alcoholic hepatitis.", HEPATOLOGY, ( OCT 2001 ) VOL. 34, NO. 4, PART 2, SUPP. [S], PP. 697A-697A. MA 2101. PUBLISHER: W B SAUNDERS CO, INDEPENDENCE SQUARE WEST CURTIS CENTER, STE 300, PHILADELPHIA, PA 19106-3399 USA. ISSN: 0270-9139., Mayo Clin, Rochester, MN USA, XP008013045 *
PATENT ABSTRACTS OF JAPAN vol. 1999, no. 13 30 November 1999 (1999-11-30) *
SPAHR L (REPRINT) ET AL: "Combination of steroids with anti- TNF alpha ( infliximab )or placebo in severe alcoholic hepatitis: Early changes in ICAM-1, histology, and biochemical parameters. A pilot study.", HEPATOLOGY, ( OCT 2001 ) VOL. 34, NO. 4, PART 2, SUPP. [S], PP. 458A-458A. MA 1143. PUBLISHER: W B SAUNDERS CO, INDEPENDENCE SQUARE WEST CURTIS CENTER, STE 300, PHILADELPHIA, PA 19106-3399 USA. ISSN: 0270-9139., Univ Hosp Geneva, Geneva, Switzerland;Essex Chem, Luzern, Switzerland, XP008013046 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9028822B2 (en) 2002-06-28 2015-05-12 Domantis Limited Antagonists against TNFR1 and methods of use therefor
WO2005117966A1 (en) * 2004-06-04 2005-12-15 Apoxis Sa Method to reduce hepatotoxicity of fas-mediated apoptosis-inducing agents

Also Published As

Publication number Publication date
AU2002343085A1 (en) 2003-06-10
GB0128138D0 (en) 2002-01-16

Similar Documents

Publication Publication Date Title
US10913792B2 (en) Treatment for rheumatoid arthritis
US5741488A (en) Treatment of rheumatoid arthritis with anti-CD4 antibodies in conjunction with anti-TNF antibodies
WO1998005357A1 (en) Anti-tnf antibodies and methotrexate in the treatment of autoimmune disease
RU2607022C2 (en) Methods and compositions for treating lupus
US20040072885A1 (en) Treatments for immune-mediated ear disorders
WO1995009652A1 (en) Treatment of autoimmune and inflammatory disorders
CA3217278A1 (en) Methods of treatment of autoimmune disorders using ilt7 binding proteins
JP2022542035A (en) Methods of treating HBV infection using anti-PRE-S1 HBV antibodies
WO2003045430A1 (en) Tnf-alpha inhibitors for the treatment of hepatic diseases
US20200062822A1 (en) Anti-tnf-alpha polypeptide composition and use thereof
CN113573732A (en) TACI-Fc fusion protein and application thereof
Gosavi et al. Comparative study on treatment of rheumatoid arthritis
JPWO2019222055A5 (en)
KR20220104789A (en) Combination of a BTK inhibitor and abatacept for the treatment of rheumatoid arthritis
Pritchard Novel therapeutic approaches to Guillain-Barré syndrome
WO2006084145A2 (en) Methods of using il-1 antagonists to reduce c-reactive protein
KR100264218B1 (en) Cdw52-specific antibody for treatment of t-cell mediated inflammation of the joints
JP7132256B2 (en) Treatment of rheumatoid arthritis
WO2022075476A1 (en) Method for treating ox40 related disease
TW202241505A (en) Method for treating autoimmune diseases and inflammation with an anti-il-17 antibody
CN117580585A (en) CD40L antagonists and their use in the treatment of lupus nephritis
JP2024516019A (en) Treatment for systemic lupus erythematosus using anti-baffr antibodies - Patents.com
AU2022342702A1 (en) Method and composition for inducing tolerance
CURE-SKŁODOWSKA ALDONA PIETRZAK, GRAŻYNA CHODOROWSKA, JANUSZ URBAN, JACEK ROLIŃSKI, BOGUSŁAW PIETRZAK
JPWO2020219960A5 (en)

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 BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE 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 NO NZ OM PH PL PT RO RU SC SD SE SG SI SK SL 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): GH GM KE LS MW MZ 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 IE IT LU MC NL PT SE 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
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP