WO2007083119A2 - Methods - Google Patents

Methods Download PDF

Info

Publication number
WO2007083119A2
WO2007083119A2 PCT/GB2007/000146 GB2007000146W WO2007083119A2 WO 2007083119 A2 WO2007083119 A2 WO 2007083119A2 GB 2007000146 W GB2007000146 W GB 2007000146W WO 2007083119 A2 WO2007083119 A2 WO 2007083119A2
Authority
WO
WIPO (PCT)
Prior art keywords
statin
cachexia
medicament
patient
previous
Prior art date
Application number
PCT/GB2007/000146
Other languages
French (fr)
Other versions
WO2007083119A3 (en
Inventor
Stephan Anker
Stephen Von Haehling
Jochen Springer
Thomas Thum
Original Assignee
Imperial Innovations Limited
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 Imperial Innovations Limited filed Critical Imperial Innovations Limited
Priority to JP2008550841A priority Critical patent/JP2009523778A/en
Priority to AU2007206776A priority patent/AU2007206776A1/en
Priority to EP07704928A priority patent/EP1973542A2/en
Publication of WO2007083119A2 publication Critical patent/WO2007083119A2/en
Publication of WO2007083119A3 publication Critical patent/WO2007083119A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/365Lactones
    • A61K31/366Lactones having six-membered rings, e.g. delta-lactones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the present invention relates to the medical use of statins for the treatment of cachexia.
  • HMG-CoA reductase inhibitors also known as 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, were originally designed to lower plasma cholesterol levels. They have revolutionized the treatment of hypercholesterolaemia. These drugs are usually well tolerated and generally safe.
  • HMG-CoA 3-hydroxy-3-methylglutaryl-coenzyme A
  • statins are currently subdivided according to their chemical structure (open-ring vj. closed-ring structure), their origin (natural vs. synthetic), and their solubility (hydrophilic vs. lipophilic), as set out in Example 1. It is interesting to note that only compounds with an open-ring structure inhibit HMG-CoA reductase. Thus, closed-ring statins undergo metabolic changes that eventually yield an open-ring structure.
  • Natural statins like mevastatin or lovastatin, are derived ' from fungal fermentation. Replacing the highly functionalized decalin ring of the fungal products with a variety of aromatic and heteroaromatic nuclei has yielded synthetic compounds, such as atorvastatin and fluvastatin. Lipophilic statins were originally thought to penetrate cell membranes more effectively than hydrophilic ones (Liao JK, J Clin Invest. 2002;l 10:285-288), however, the evidence suggests otherwise.
  • pleiotropic effects include improvement of endothelial dysfunction, release of endothelial progenitor cells, anti-inflammatory properties, and a number of anti-tumour activities.
  • Cachexia (unintentional weight loss) is a devastating disorder with a poor prognosis. It is seen as a consequence of many chronic diseases, including acquired immune deficiency syndrome (AIDS), liver cirrhosis, chronic obstructive pulmonary disease (COPD), chronic renal failure (ie chronic kidney disease), chronic infections including pneumonia, cancer (cancer cachexia), diabetes and heart disease including hypertension and chronic heart failure (CHF) (cardiac cachexia). Cachexia may also occur idiopathically. Cachexia is also associated with ageing, rheumatoid arthritis, diabetes, stroke, burns, trauma and other acute injury, sepsis, HIV/AIDS and other infections.
  • AIDS acquired immune deficiency syndrome
  • COPD chronic obstructive pulmonary disease
  • COPD chronic renal failure
  • chronic kidney disease chronic infections including pneumonia
  • cancer cancer
  • diabetes and heart disease including hypertension and chronic heart failure (CHF) (cardiac cachexia).
  • Cachexia may also occur idiopathically. Cachexia is also
  • TNF ⁇ interleukin 1
  • IL-6 interleukin-6
  • TNF ⁇ tumour necrosis factor- ⁇
  • TNF ⁇ plays a considerable role in chronic heart failure and its progression to cardiac cachexia, as this cytokine is involved in endothelial dysfunction and left ventricular impairment (von Haehling et al Basic Res Cardiol 2004;99: 18-28).
  • High plasma levels of TNF ⁇ and its soluble receptors TNFR-I and TNFR-2 predict a poor survival (Deswal et al Circulation 2001;103:2055-2059; Rauchhaus et al Circulation 2000 19; 102:3060-3067).
  • statins may inhibit the production of such proinflammatory substances.
  • Lovastatin for example, was found to inhibit the induction of TNFa 5 IL-I 5 and IL-6 in certain rat cell lines (Pahan et al J Clin Invest 1997;100:2671-2679).
  • CRP C-reactive protein
  • CRP is much more than a mere marker of the body's inflammatory load.
  • CRP was shown to activate endothelial cells, which, in turn, express Intracellular Adhesion Molecule- 1 (ICAM-I) (Pasceri et al Circulation. 2000;102:2165-2168).
  • IAM-I Intracellular Adhesion Molecule- 1
  • VCAM-I vascular-cell adhesion molecule-1
  • E-selectin E-selectin.
  • CRP monocyte chemotactic protein- 1
  • MCP-I monocyte chemotactic protein- 1
  • PRNCE Pravastatin Inflammation/CRP Evaluation
  • Statins also appear to reduce the number of inflammatory cells in atherosclerotic plaques. This observation is in line with reports that showed a statin-mediated reduction in the number of adhesion sites on the cell surfaces of leukocytes.
  • a study in isolated human monocytes recently found that both pravastatin and fluvastatin inhibited the expression of ICAM-I on these cells (Takahashi et al J Leukoc Biol 2005:77:400-407).
  • IFN- ⁇ interferon- ⁇
  • Rho proteins form a family of proteins that activate various kinases. Extracellular stimuli convert the inactive form of Rho (GDP -Rho) to its active form (GTP -Rho). In fact, efficient leukocyte extravasation requires Rho signaling not only within the migrating leukocytes but also within the endothelial lining of the vessel wall (Strey et al FEBS Lett 2002;517:261-266). Moreover, Rho mediates dynamic reorganization of cytoskeletal proteins, such as stress fibre and focal adhesion formation (Amano et al Exp Cell Res 2000;261:44-51). Thus, Rho is believed to play an important role in local inflammatory responses.
  • Endothelial dysfunction is frequently seen in cachectic patients.
  • Statins have been shown to improve endothelial dysfunction by a number of mechanisms, to possess anti-inflammatory properties, and to have various anti-tumour effects (the latter applies mostly for lovastatin, which has been studied in greatest detail).
  • pleiotropic effects of statins appear independent of their cholesterol- lowering features.
  • cholesterol reduction itself does not yield, for example, beneficial effects on endothelial function.
  • Pleiotropic effects appear to be dose-dependent, and it is likely that pleiotropic effects appear even without cholesterol reduction, i.e. at doses that are so low that no cholesterol reduction is seen.
  • Achieving pleiotropic effects without cholesterol reduction might be important, as a number of diseases do have better outcomes with high cholesterol levels (e.g. chronic heart failure, some types of cancer).
  • Very low doses of statins may therefore exert pleiotropic effects without cholesterol reduction. In this sense, cholesterol reduction would merely be a side effect.
  • simvastatin (20 mg/kg per day) negatively affected the wasting pattern induced by Yoshida AH-130 ascites hepatoma in rats.
  • the organ weights of liver, spleen, heart, gastrocnemius, and soleus decreased by four days of simvastatin treatment.
  • the administration of simvastatin to control animals (without hepatoma) yielded no effects with the exception of a significant reduction in heart weight (controls without simvastatin: 501 ⁇ 58 mg, controls with simvastatin: 426 ⁇ 65 mg, p ⁇ 0.05).
  • statin the effect of the statin on the rats was measured after only 4 days. This duration was not even able to affect cholesterol/triglyceride levels, although HDL levels were increased. Moreover, the amount of statin used in this study may not have been the correct quantity.
  • statin can be used to treat cachexia.
  • a first aspect of the invention provides the use of at least one statin in the manufacture of a medicament for the prevention or treatment of cachexia in a patient, wherein the patient is human or a domestic or farm animal.
  • statin can be used to treat cachexia.
  • patient includes a human patient and also domestic animals such as cats and dogs, and farm animals such as cows, pigs, horses, sheep, goats and the like.
  • the patient is not a rat. It is preferred that the patient is human.
  • the medicament of the first aspect of the invention may be used to treat a patient diagnosed with cachexia, and to treat a patient at risk of developing cachexia.
  • the patient has been diagnosed as having cachexia.
  • Cachexia can be defined as a history of weight loss, for example >5% (preferably >6%) weight loss over a period of at least six months. However, in patients with cancer and aging at least 4% weight loss over three months can be used as a basis to diagnose cachexia. Cachexia can also be defined as a low body mass index, for example ⁇ 20, ⁇ 21, ⁇ 21.5, ⁇ 22 kg/m 2 over a period of time, for example six months.
  • Diagnosis can be made by asking the patient for their weight over the preceding months.
  • the weight of an individual can be monitored over a period of time, for example six months.
  • the medicament may comprise an appropriate quantity of statin to treat cachexia.
  • Existing dosage regimes for human patients are provided in the accompanying examples.
  • statin may induce beneficial pleiotropic effects on the patient without substantially reducing cholesterol level.
  • an embodiment of this aspect of the invention is wherein the medicament is for administering an amount of statin that does not substantially reduce cholesterol level.
  • statin may induce beneficial pleiotropic effects on the patient. These pleiotropic effects may be of benefit for the treatment of cachexia. Therefore the medicament may be for administering an amount of statin that induces pleiotropic effects on the patient. This amount of statin may also not substantially reduce cholesterol levels in a patient treated with the medicament.
  • pleiotropic effects we include improvement of endothelial dysfunction, release of endothelial progenitor cells, anti-inflammatory properties, and a number of anti-tumour activities.
  • not substantially reduce cholesterol level we include that the cholesterol level of a patient treated with the medicament is substantially similar to the cholesterol level of a patient who has not been treated with the medicament. We also include where the cholesterol level of a patient treated with the medicament is substantially similar to the cholesterol level of the same patient before administration of the medicament. We also include where the cholesterol levels of a population of patients treated with the medicament is substantially similar to the cholesterol levels of a population of patients who has not been treated with the medicament. In each of these cases, the medicament can be considered not to have substantially reduced cholesterol levels.
  • the cholesterol level is considered to be "substantially similar” if it is at least 90% of the reference cholesterol levels, preferably, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 101%, 102%, 103%, 104%, 105%, 106%, 107%, 108%, 109%, 110% of those levels.
  • statin we include atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, cerivastatin, mevastatin or pravastatin.
  • statin is simvastatin or atorvastatin.
  • Statins are well known compounds and may be obtained from a number of sources. For example, rosuvastatin (AstraZeneca); atorvastatin (Pfizer); simvastatin (Merck); fluvastatin (Novartis); pravastatin (Bristol-Myers Squibb); lovastatin (Merck), pitvastatin (Kowa).
  • the medicament comprises a daily unit dose of less than 5mg of rosuvastatin or simvastatin per day; or less than 10 mg of atorvastatin, lovastatin or pravastatin per day; or less than 20 mg of fluvastatin per day.
  • less than 5 mg we include between 0.01 mg and 4.9 mg per day, for example less than or equal to 4.9, 4.8, 4.7, 4.6 4.5, 4, 3.5, 3, 2.5, 2, 1.5, 1, 0.5, 0.1, 0.05, 0.04, 0.03, 0.02 or 0.01 mg per day.
  • less than 10 mg we include between 0.01 mg and 9.9 mg per day, for example less than or equal to 9.9, 9.8, 9.7, 9.6, 9.5, 9, 8.5, 8, 7.5, 7, 6.5, 6, 5.5, 5, 4.5, 4, 3.5, 3, 2.5, 2, 1.5, 1, 0.5, 0.1, 0.05, 0.04, 0.03, 0.02 or 0.01 mg per day.
  • nig between 0.01 mg and 19.9 nig per day, for example we include less than or equal to 19.9, 19.8, 19.7, 19.6, 19.5, 19, 18.5, 18, 17.5, 17, 16.5, 16, 15.5, 15, 14.5, 14, 13.5, 13, 12.5, 12, 11.5, 11, 10.5, 10, 9.5, 9, 8.5, 8, 7.5, 7, 6.5, 6, 5.5, 5, 4.5, 4, 3.5, 3, 2.5, 2, 1.5, 1, 0.5, 0.1, 0.05, 0.04, 0.03, 0.02 or 0.01 mg per day.
  • the medicament may comprise a quantity of the statin equivalent to the daily unit dose.
  • the medicament may comprise a slow release formulation of the statin having the property of releasing the daily unit dose.
  • Cachexia can occur in patients with a wide variety of diseases including infectious diseases such as acquired immune deficiency syndrome (AIDS), liver cirrhosis, chronic obstructive pulmonary disease (COPD), chronic renal failure, chronic infections including pneumonia, cancer (cancer cachexia), diabetes mellitus and heart disease including hypertension and chronic heart failure (CHF) (cardiac cachexia). Cachexia may also occur idiopathically. Cachexia is also associated with ageing, rheumatoid arthritis, diabetes, stroke, burns, trauma and other acute injury, sepsis, HIV/ AIDS and other infections.
  • infectious diseases such as acquired immune deficiency syndrome (AIDS), liver cirrhosis, chronic obstructive pulmonary disease (COPD), chronic renal failure, chronic infections including pneumonia, cancer (cancer cachexia), diabetes mellitus and heart disease including hypertension and chronic heart failure (CHF) (cardiac cachexia). Cachexia may also occur idiopathically. Cachexia is also associated with ageing, rheumatoi
  • the cachexia is cancer cachexia, cardiac cachexia (for example cachexia due to CHF or cardiomyopathy) or chronic obstructive pulmonary disease (COPD) cachexia.
  • cardiac cachexia for example cachexia due to CHF or cardiomyopathy
  • COPD chronic obstructive pulmonary disease
  • the cachexia may instead be, for example, cachexia associated with liver cirrhosis, chronic renal failure, rheumatoid arthritis, diabetes, stroke, burns, trauma or other acute injury, sepsis, HIV/ AIDS or other infections or ageing.
  • the medicament or method of treatment
  • the statin used in this aspect of the invention has been prepared using the micro-crystallisation method disclosed in WO 03/103640 (herein incorporated by reference).
  • micro crystallization can improve drug activity and final product characteristics. Poor water solubility (including simvastatin, atorvastatin, and lovastatin; fluvastatin and cerivastatin) correlates with slow dissolution rate, and decreasing particle size increases the surface area, which leads to an increase in dissolution rate. This can be accomplished using micro crystallization, because such particles are small particles of drug substance, typically less than 1000 nanometers (nm) in diameter, which are produced by milling the drug substance using a wet milling technique.
  • micro crystallized particles of the drug are stabilized against agglomeration by surface adsorption of selected stabilizers.
  • the result is an aqueous dispersion of the drug substance that behaves like a solution-a NanoCrystal colloidal dispersion, which can be processed into finished dosage forms for all routes of administration.
  • any statin might be treated in such a way, however, the oral bioavailability of pravastatin and rosuvastatin (which are hydrophilic) appears to be less likely to benefit from such treatment.
  • the lipophilic drugs cerivastatin and pravastatin have per se a high bioavailability (of approximately 60%) which could be enhanced by micro crystallization.
  • statin will normally be administered orally or by any parenteral route, in the form of a pharmaceutical formulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage form.
  • the medicament may be administered at varying doses.
  • statin can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
  • statin can be administered orally, buccally or sublingually in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed- or controlled- release applications.
  • the stain may also be administered via intracavernosal injection.
  • Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropyhnethylcellulose (HPMC), hydroxy-propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
  • excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine
  • disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates,
  • Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols.
  • the statin may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • the statin can also be administered parenterally, for example, intravenously, intra- arterially, intraperitoneally, intrathecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • suitable parenteral formulations under sterile conditions is readily accomplished ⁇ by standard pharmaceutical techniques well-known to those skilled in the art.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • the statin can also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant.
  • a suitable propellant e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA 134A3 or 1,1,1,2,3,3,3-heptafluoro ⁇ ropane (HFA 227EA3), carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active compound, e.g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e.g. sorbitan trioleate.
  • a lubricant e.g. sorbitan trioleate.
  • Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a statin and a suitable powder base such as lactose or starch.
  • Aerosol or dry powder formulations are preferably arranged so that each metered dose or "puff contains precise quantity of statin for delivery to the patient. It will be appreciated that he overall daily dose with an aerosol will vary from patient to patient, and may be administered in a single dose or, more usually, in divided- doses throughout the day.
  • statin can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder.
  • the statin may also be transdermally administered, for example, by the use of a skin patch. They may also be administered by the ocular route, particularly for treating diseases of the eye.
  • the statin can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.
  • the statin can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water.
  • they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia," and mouth-washes comprising the active ingredient in a suitable liquid carrier.
  • statin is the preferred route, being the most convenient.
  • the drug may be administered parenterally, e.g. sublingually or buccally.
  • statin For oral and parenteral administration to human patients, the daily dosage level of statin are provided above.
  • the physician in any event will determine the actual dosage which will be most suitable for any individual patient and it will vary with the age, weight and response of the particular patient.
  • the above dosages are exemplary of the average case. There can, of course, be individual instances where higher or lower dosage ranges are merited and such are within the scope of this invention.
  • the medicament can be used for the prevention or treatment of cachexia.
  • Existing treatment for this disorder include the following therapeutic agents: growth hormone, anabolic steroids, and appetite stimulants for some subgroups of cachexia.
  • the medicament comprises one or more of such therapeutic agents for preventing or treating cachexia.
  • a further aspect of the invention provide a method of treating a patient with or at risk of developing cachexia comprising administering an appropriate quantity of at least one statin, wherein the patient is human or a domestic or farm animal.
  • the patient is human.
  • An embodiment of this aspect of the invention is wherein the patient is administered an amount of statin that does not substantially reduce cholesterol level.
  • statin is atorvastatin, fmvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, cerivastatin, mevastatin or pitavastatin.
  • statin is simvastatin or atorvastatin.
  • the method uses a quantity of the statin equivalent to a daily unit dose.
  • An embodiment of this aspect of the invention is wherein the method uses a slow release formulation of the statin having the property of releasing a daily unit dose.
  • An embodiment of this aspect of the invention is wherein the statin has been prepared using micro-crystallisation.
  • An embodiment of this aspect of the invention is wherein the method further comprises using one or more further therapeutic agents for preventing or treating cachexia.
  • An embodiment of this aspect of the invention is wherein the cachexia is cancer cachexia, cardiac cachexia or chronic obstructive pulmonary disease (COPD) cachexia.
  • the cachexia is cancer cachexia, cardiac cachexia or chronic obstructive pulmonary disease (COPD) cachexia.
  • COPD chronic obstructive pulmonary disease
  • Example 2 shows a higher likelihood of experiencing weight gain (assessed as weight gain of more than 5%) for patients treated with statins.
  • Example 3 also shows a positive trend for low dose statin on body weight.
  • the medicament or method is considered to be useful in assisting increase in lean tissue or skeletal muscle mass.
  • Example 3 shows a positive trend for low dose statin on lean tissue and skeletal muscle mass.
  • Muscle mass can be measured in humans in several ways, as will be well known to the person skilled in the art, for example: - antropometry (using calipers to assess skin fold thickness and then estimate muscle mass)
  • DEXA scanning (dual X-ray absorptiometry, as described in, for example Anker et al (1999) 20, 683-693).
  • DEXA is the method most often used in cachexia research projects
  • the skeletal muscle mass increase may be relative to the starting point of muscle mass in the patient or as the proportion of the patient's body mass. From a muscle mass and strength point of view either is desirable. Preferably after treatment there is more muscle than before: whether this is accompanied by an increase in body weight or not is of lesser importance for muscle anabolic interventions. It is preferable that muscle increases without loss of fat, i.e. that muscle gain leads to overall weight gain.
  • FIG. 2 Pathway of cholesterol biosynthesis.
  • Statins block the rate-limiting step in the cascade by inhibition of HMG-CoA reductase. The inhibition can be overcome by addition of mevalonate in experimental models.
  • CoA - coenzyme A HMG- 3-hydroxy-3-methylglutaryl, PP - pyrophosphate.
  • Figure 3 Overview of the principal pleiotropic effects of statins on (A) the endothelium, (B) the immune system, and (C) tumour development and metastasis.
  • statins have been shown to improve endothelial function by increasing nitric oxide (NO) production and release. Statins also increase the number of endothelial progenitor cells in the bloodstream.
  • NO nitric oxide
  • proinflammatory mediators e.g. TNF-a, IL-I, IL-6
  • adhesion molecule expression e.g. ICAM-I
  • statins The release of C-reactive protein (CRP) has been proven in several clinical settings.
  • Anti-tumour activities of statins comprise the induction of apoptosis, growth arrest by inhibiting the cell cycle of malignant cells, and the reduction of the invasiveness of malignant cells at secondary sites.
  • the following table includes information on the characteristics of statins that may be used in the invention.
  • ** C max may vary according to the administered dose and the patient population investigated.
  • Example 2 Use of statins for the treatment of cachexia
  • statins used in the ELITE 2 trial were:
  • RESULTS ELITE 2 study. Cox Proportional Hazard Analysis of the impact of being on statin at baseline (BL) on the subsequent development of weight gain >5%. The impact is highly significant (p ⁇ 0.0001) and independent of the severity of heart failure as measured by LVEF, NYHA class, clinical oedema status, degree of kidney dysfunction (i.e. creatinine [crea] levels) and heart failure aetiology. In 3030 patients all information for this analysis was available.
  • Model Proportional Hazards. Row exclusion: ELITE2 11- 05B-w-change-080805.
  • ELITE 2 study - the subgroup of patients with a diagnosis of chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • statin therapy was associated with weight gain only in patients with COPD (33.4% increase in the event rate of weight gain), unlike the general population of the ELITE 2 trial in which the occurrence of weight gain due to statin therapy was 17% reduced (see Table 2).
  • the animals were seen at least twice per day to ensure precise survival data.
  • Body Weight was monitored every two days throughout the entire study period.
  • Body Composition was measured the day before tumour inoculation and in two day intervals after that until the end of the study using NMR-scans (EchoMRI systems). Read-out is fat, lean mass, total water and free water. Locomotor Activity Assessed using Supermex system over a period of 24 hours in cages where the animals are placed individually (type 3 cage).
  • Echocardiography was performed in all animals on day 0 and on day 12 using a high resolution echocardiography system (Vevo770, Visual Sonics) to determine the following parameters:
  • LVEF left ventricular ejection fraction
  • LVFS left ventricular fractional shortening
  • LVEDD and LVESD left ventricular end-diasolic and end-systolic diameters
  • M-Mode septum and inferior wall thickness
  • rVSd and IVSs thickness of the inter- ventricular septum in diastole and systole diastolic filling
  • Heart function Heart function was assessed by high resolution echocardiography using the Vevo 770 system (Visualsonics, Toronto, Canada).
  • the left ventricular (LV) ejection fraction, LV fractional shortening, LV end-diastolic diameter, LV end-systolic diameter, LV posterior wall thickness in diastole and systole, thickness of the inter-ventricular septum in diastole and systole, the LV stroke volume and the diastolic function (E/ A) were assessed.
  • the results showed neutral results for all parameter assessed (ANOVA, all p>0.4).
  • Fat mass did not show any significant difference between the simvastatin groups and the placebo group.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Diabetes (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Obesity (AREA)
  • Urology & Nephrology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Hematology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Endocrinology (AREA)
  • Vascular Medicine (AREA)
  • Immunology (AREA)
  • Emergency Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Dermatology (AREA)
  • Neurology (AREA)
  • Pulmonology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The use of at least one statin in the manufacture of a medicament for the prevention or treatment of cachexia in a patient, wherein the patient is human or a domestic or farm animal

Description

Methods
The present invention relates to the medical use of statins for the treatment of cachexia.
Statins, also known as 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, were originally designed to lower plasma cholesterol levels. They have revolutionized the treatment of hypercholesterolaemia. These drugs are usually well tolerated and generally safe.
A number of different statin classifications have been suggested. In essence, statins are currently subdivided according to their chemical structure (open-ring vj. closed-ring structure), their origin (natural vs. synthetic), and their solubility (hydrophilic vs. lipophilic), as set out in Example 1. It is interesting to note that only compounds with an open-ring structure inhibit HMG-CoA reductase. Thus, closed-ring statins undergo metabolic changes that eventually yield an open-ring structure.
Natural statins, like mevastatin or lovastatin, are derived' from fungal fermentation. Replacing the highly functionalized decalin ring of the fungal products with a variety of aromatic and heteroaromatic nuclei has yielded synthetic compounds, such as atorvastatin and fluvastatin. Lipophilic statins were originally thought to penetrate cell membranes more effectively than hydrophilic ones (Liao JK, J Clin Invest. 2002;l 10:285-288), however, the evidence suggests otherwise.
Effects beyond mere cholesterol reduction, the so-called pleiotropic effects, have recently received tremendous attention. These pleiotropic effects include improvement of endothelial dysfunction, release of endothelial progenitor cells, anti-inflammatory properties, and a number of anti-tumour activities.
Recent work on statins has largely focused on these pleiotropic effects. Promising results suggest beneficial effects in patients with multiple sclerosis (Sena et al, J Neurol 2003;250:754-755; Vollmer et al, Lancet 2004;363:1607-1608), Alzheimer's disease (Jick et al, Lancet 2000;356: 1627-1631), osteoporosis (Pasco et al Arch Intern Med 2002;162:537-540), age-related macular degeneration (with mixed results) (McGwin et al Br J Ophthalmol 2003 Sep;87:l 121-1125; Klein et al Arch Ophthalmol 2003;121:1151-1155). some types of cancer (Graaf et al Cancer Treat Rev. 2004 Nov;30(7):609-641), and heart failure (Horwich et al J Am Coll Cardiol. 2004;43: 642-648; Anker et al Circulation 2002; 106 Suppl 11:2535 (Abstract)).
Cachexia (unintentional weight loss) is a devastating disorder with a poor prognosis. It is seen as a consequence of many chronic diseases, including acquired immune deficiency syndrome (AIDS), liver cirrhosis, chronic obstructive pulmonary disease (COPD), chronic renal failure (ie chronic kidney disease), chronic infections including pneumonia, cancer (cancer cachexia), diabetes and heart disease including hypertension and chronic heart failure (CHF) (cardiac cachexia). Cachexia may also occur idiopathically. Cachexia is also associated with ageing, rheumatoid arthritis, diabetes, stroke, burns, trauma and other acute injury, sepsis, HIV/AIDS and other infections.
Although a final common pathway of all forms of cachexia has as yet not been established, it is thought that pro-inflammatory cytokines play a crucial role in. this sense. For example, TNF α, interleukin (IL) 1, and IL-6 are known to stimulate the ubiquitin-proteasome pathway in skeletal muscle (Zamir et al Arch Surg 1992;127:170-174; Zamir et al Metabolism 1993;42:204-208; Goodman, Proc Soc Exp Biol Med 1994;205 : 182-185). Among these substances, tumour necrosis factor-α (TNFα) has been considerably researched during the last several years. TNFα plays a considerable role in chronic heart failure and its progression to cardiac cachexia, as this cytokine is involved in endothelial dysfunction and left ventricular impairment (von Haehling et al Basic Res Cardiol 2004;99: 18-28). High plasma levels of TNFα and its soluble receptors TNFR-I and TNFR-2 predict a poor survival (Deswal et al Circulation 2001;103:2055-2059; Rauchhaus et al Circulation 2000 19; 102:3060-3067). Several studies have shown that statins may inhibit the production of such proinflammatory substances. Lovastatin, for example, was found to inhibit the induction of TNFa5 IL-I5 and IL-6 in certain rat cell lines (Pahan et al J Clin Invest 1997;100:2671-2679). In a study in 40 hypercholesterolaemic patients, pravastatin 40 mg once daily led to a significant reduction in TNFα plasma levels after eight weeks of treatment as compared to placebo (pravastatin 1.10, placebo 1.33 ρg/ml, ρ=0.032) (Solheim et al Atherosclerosis 2001,157:411-415). This effect was most pronounced in the subgroup of smokers. IL-6 plasma values were left unaffected. Another study found a significant decrease in TNFα release into the supernatant of phorbol myristate acetate (PMA) stimulated human monocytes after the addition of increasing doses of pravastatin as compared to PMA treated samples (p<0.01) (Grip et al Eur J Pharmacol 2000;410:83-92). Pravastatin had no effect on PMA induced IL-6 production. Data from large-scale clinical studies are currently not available.
Furthermore, C-reactive protein (CRP) levels are an independent predictor of future coronary events (Ridker et al N Engl J Med. 1997,336:973-979). Its plasma values are positively correlated with weight loss, the occurrence of cachexia, and recurrence in advanced cancer (Mahmoud and Rivera, Curr Oncol Rep 2002;4:250-255). Its role as a predictor of survival has been shown in multiple myeloma, melanoma, lymphoma, ovarian, renal, pancreatic, and gastrointestinal tumours (Mahmoud and Rivera, Curr Oncol Rep 2002;4:250-255). Recent studies suggest that CRP is much more than a mere marker of the body's inflammatory load. In cultured human umbilical vein endothelial cells, CRP was shown to activate endothelial cells, which, in turn, express Intracellular Adhesion Molecule- 1 (ICAM-I) (Pasceri et al Circulation. 2000;102:2165-2168). CRP also induces other adhesion molecules in endothelial cells such as vascular-cell adhesion molecule-1 (VCAM-I) and E-selectin. These molecules are involved in leukocyte- binding to the endothelial layer. CRP also activates the expression of monocyte chemotactic protein- 1 (MCP-I) (Pasceri et al Circulation. 2001; 103: 2531-2534). Several recent large-scale studies have shown that plasma CRP levels can be reduced with statin therapy. The Pravastatin Inflammation/CRP Evaluation (PRINCE) study demonstrated a significant CRP reduction in 1182 patients with a history of myocardial infarction, stroke, or arterial revascularisation procedure using pravastatin 40 mg once daily (Albert et al JAMA 2001;286:64-70). The reduction was 13,1% as compared to baseline. A re-analysis of the Pravastatin or Atorvastatin Evaluation and Infection Therapy - Thrombolysis in Myocardial Infarction 22 (PROVE IT-TIMI 22) database found recently that both drugs reduce CRP levels within 30 days of initiation significantly among 3,745 patients with acute coronary syndromes (Ridker et al N Engl J Med 2005;352:20-28). Atorvastatin 80 mg once daily was more effective than pravastatin 40 mg once daily at 30 days (median CRP level: 1.6 vs. 2.3 mg/1, pO.001) and at four months (1.3 vs. 2.1 mg/1, p<0.001). It is interesting to note that the achieved CRP and LDL levels were independent of each other (Ridker et al N Engl J Med 2005;352:20-28).
Statins also appear to reduce the number of inflammatory cells in atherosclerotic plaques. This observation is in line with reports that showed a statin-mediated reduction in the number of adhesion sites on the cell surfaces of leukocytes. A study in isolated human monocytes recently found that both pravastatin and fluvastatin inhibited the expression of ICAM-I on these cells (Takahashi et al J Leukoc Biol 2005:77:400-407). In this study, the release of TNFα and interferon-γ (IFN-γ) was inhibited by both statins only in the presence of IL-18 (formerly known as IFN-γ-inducing factor). Another study described a statin-mediated reduction of the expression of a counterreceptor of ICAM-I on leukocytes (Weitz- Schmidt G et al Nat Med 2001; 7: 687 - 692). This down-regulation reduced their adhesion to endothelial cells (Weitz-Schmidt G et al Nat Med 2001; 7: 687 - 692). These results are consistent with reports in which simvastatin pretreatment inhibited Staphylococcus aureus-in&ac&ά leukocyte rolling and adherence in the rat mesenteric circulation as assessed using intravital microscopy (Pruefer et al Circulation 2002; 106:2104-2110). Leukocyte transmigration was also significantly decreased by such treatment. Some of these statin-mediated effects on immunological responses might be regulated by Rho proteins. These form a family of proteins that activate various kinases. Extracellular stimuli convert the inactive form of Rho (GDP -Rho) to its active form (GTP -Rho). In fact, efficient leukocyte extravasation requires Rho signaling not only within the migrating leukocytes but also within the endothelial lining of the vessel wall (Strey et al FEBS Lett 2002;517:261-266). Moreover, Rho mediates dynamic reorganization of cytoskeletal proteins, such as stress fibre and focal adhesion formation (Amano et al Exp Cell Res 2000;261:44-51). Thus, Rho is believed to play an important role in local inflammatory responses.
Endothelial dysfunction is frequently seen in cachectic patients. Statins have been shown to improve endothelial dysfunction by a number of mechanisms, to possess anti-inflammatory properties, and to have various anti-tumour effects (the latter applies mostly for lovastatin, which has been studied in greatest detail).
The so-called pleiotropic effects of statins appear independent of their cholesterol- lowering features. In fact, cholesterol reduction itself does not yield, for example, beneficial effects on endothelial function. Pleiotropic effects, however, appear to be dose-dependent, and it is likely that pleiotropic effects appear even without cholesterol reduction, i.e. at doses that are so low that no cholesterol reduction is seen. At present clinical studies to support this view are missing. Achieving pleiotropic effects without cholesterol reduction might be important, as a number of diseases do have better outcomes with high cholesterol levels (e.g. chronic heart failure, some types of cancer). Very low doses of statins may therefore exert pleiotropic effects without cholesterol reduction. In this sense, cholesterol reduction would merely be a side effect.
The only study in an animal model of cancer cachexia ended in disappointment (Muscaritoli et al Nutrition 2003;19:936-939). In essence, simvastatin (20 mg/kg per day) negatively affected the wasting pattern induced by Yoshida AH-130 ascites hepatoma in rats. The organ weights of liver, spleen, heart, gastrocnemius, and soleus decreased by four days of simvastatin treatment. The administration of simvastatin to control animals (without hepatoma) yielded no effects with the exception of a significant reduction in heart weight (controls without simvastatin: 501±58 mg, controls with simvastatin: 426±65 mg, p<0.05).
However, the effect of the statin on the rats was measured after only 4 days. This duration was not even able to affect cholesterol/triglyceride levels, although HDL levels were increased. Moreover, the amount of statin used in this study may not have been the correct quantity.
We have now determined that statin can be used to treat cachexia.
The listing or discussion of a prior-published document in this specification should not necessarily be taken as an acknowledgement that the document is part of the state of the art or is common general knowledge.
A first aspect of the invention provides the use of at least one statin in the manufacture of a medicament for the prevention or treatment of cachexia in a patient, wherein the patient is human or a domestic or farm animal.
As shown in the accompanying examples, contrary to the finding of Muscaritoli et al supra we have determined that statin can be used to treat cachexia.
The term "patient" includes a human patient and also domestic animals such as cats and dogs, and farm animals such as cows, pigs, horses, sheep, goats and the like. The patient is not a rat. It is preferred that the patient is human.
The medicament of the first aspect of the invention may be used to treat a patient diagnosed with cachexia, and to treat a patient at risk of developing cachexia. Preferably the patient has been diagnosed as having cachexia.
Cachexia can be defined as a history of weight loss, for example >5% (preferably >6%) weight loss over a period of at least six months. However, in patients with cancer and aging at least 4% weight loss over three months can be used as a basis to diagnose cachexia. Cachexia can also be defined as a low body mass index, for example <20, <21, <21.5, <22 kg/m2 over a period of time, for example six months.
Diagnosis can be made by asking the patient for their weight over the preceding months. Alternatively, the weight of an individual can be monitored over a period of time, for example six months.
The medicament may comprise an appropriate quantity of statin to treat cachexia. Existing dosage regimes for human patients are provided in the accompanying examples.
However, as discussed above, low amounts of statin may induce beneficial pleiotropic effects on the patient without substantially reducing cholesterol level.
Accordingly, an embodiment of this aspect of the invention is wherein the medicament is for administering an amount of statin that does not substantially reduce cholesterol level.
As discussed above, low amounts of statin may induce beneficial pleiotropic effects on the patient. These pleiotropic effects may be of benefit for the treatment of cachexia. Therefore the medicament may be for administering an amount of statin that induces pleiotropic effects on the patient. This amount of statin may also not substantially reduce cholesterol levels in a patient treated with the medicament.
By "pleiotropic effects" we include improvement of endothelial dysfunction, release of endothelial progenitor cells, anti-inflammatory properties, and a number of anti-tumour activities.
By "not substantially reduce cholesterol level" we include that the cholesterol level of a patient treated with the medicament is substantially similar to the cholesterol level of a patient who has not been treated with the medicament. We also include where the cholesterol level of a patient treated with the medicament is substantially similar to the cholesterol level of the same patient before administration of the medicament. We also include where the cholesterol levels of a population of patients treated with the medicament is substantially similar to the cholesterol levels of a population of patients who has not been treated with the medicament. In each of these cases, the medicament can be considered not to have substantially reduced cholesterol levels.
In each of the above cases, the cholesterol level is considered to be "substantially similar" if it is at least 90% of the reference cholesterol levels, preferably, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 100%, 101%, 102%, 103%, 104%, 105%, 106%, 107%, 108%, 109%, 110% of those levels.
By "statin" we include atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, cerivastatin, mevastatin or pravastatin. Preferably the statin is simvastatin or atorvastatin.
Statins are well known compounds and may be obtained from a number of sources. For example, rosuvastatin (AstraZeneca); atorvastatin (Pfizer); simvastatin (Merck); fluvastatin (Novartis); pravastatin (Bristol-Myers Squibb); lovastatin (Merck), pitvastatin (Kowa).
It is preferred that the medicament comprises a daily unit dose of less than 5mg of rosuvastatin or simvastatin per day; or less than 10 mg of atorvastatin, lovastatin or pravastatin per day; or less than 20 mg of fluvastatin per day.
By "less than 5 mg" we include between 0.01 mg and 4.9 mg per day, for example less than or equal to 4.9, 4.8, 4.7, 4.6 4.5, 4, 3.5, 3, 2.5, 2, 1.5, 1, 0.5, 0.1, 0.05, 0.04, 0.03, 0.02 or 0.01 mg per day.
By "less than 10 mg" we include between 0.01 mg and 9.9 mg per day, for example less than or equal to 9.9, 9.8, 9.7, 9.6, 9.5, 9, 8.5, 8, 7.5, 7, 6.5, 6, 5.5, 5, 4.5, 4, 3.5, 3, 2.5, 2, 1.5, 1, 0.5, 0.1, 0.05, 0.04, 0.03, 0.02 or 0.01 mg per day. By "less than 20 nig" between 0.01 mg and 19.9 nig per day, for example we include less than or equal to 19.9, 19.8, 19.7, 19.6, 19.5, 19, 18.5, 18, 17.5, 17, 16.5, 16, 15.5, 15, 14.5, 14, 13.5, 13, 12.5, 12, 11.5, 11, 10.5, 10, 9.5, 9, 8.5, 8, 7.5, 7, 6.5, 6, 5.5, 5, 4.5, 4, 3.5, 3, 2.5, 2, 1.5, 1, 0.5, 0.1, 0.05, 0.04, 0.03, 0.02 or 0.01 mg per day.
The medicament may comprise a quantity of the statin equivalent to the daily unit dose. Alternatively, the medicament may comprise a slow release formulation of the statin having the property of releasing the daily unit dose.
As mentioned above, "cachexia" can occur in patients with a wide variety of diseases including infectious diseases such as acquired immune deficiency syndrome (AIDS), liver cirrhosis, chronic obstructive pulmonary disease (COPD), chronic renal failure, chronic infections including pneumonia, cancer (cancer cachexia), diabetes mellitus and heart disease including hypertension and chronic heart failure (CHF) (cardiac cachexia). Cachexia may also occur idiopathically. Cachexia is also associated with ageing, rheumatoid arthritis, diabetes, stroke, burns, trauma and other acute injury, sepsis, HIV/ AIDS and other infections.
Preferably, the cachexia is cancer cachexia, cardiac cachexia (for example cachexia due to CHF or cardiomyopathy) or chronic obstructive pulmonary disease (COPD) cachexia.
The cachexia may instead be, for example, cachexia associated with liver cirrhosis, chronic renal failure, rheumatoid arthritis, diabetes, stroke, burns, trauma or other acute injury, sepsis, HIV/ AIDS or other infections or ageing. Thus, the medicament (or method of treatment) may be for treating patients selected as above 60, 65, 70 or 75 years of age. Preferably, the statin used in this aspect of the invention has been prepared using the micro-crystallisation method disclosed in WO 03/103640 (herein incorporated by reference).
The procedure of micro crystallization (NanoCrystal Technology) can improve drug activity and final product characteristics. Poor water solubility (including simvastatin, atorvastatin, and lovastatin; fluvastatin and cerivastatin) correlates with slow dissolution rate, and decreasing particle size increases the surface area, which leads to an increase in dissolution rate. This can be accomplished using micro crystallization, because such particles are small particles of drug substance, typically less than 1000 nanometers (nm) in diameter, which are produced by milling the drug substance using a wet milling technique.
The micro crystallized particles of the drug are stabilized against agglomeration by surface adsorption of selected stabilizers. The result is an aqueous dispersion of the drug substance that behaves like a solution-a NanoCrystal colloidal dispersion, which can be processed into finished dosage forms for all routes of administration.
This has been shown to be particularly effective at enhancing oral bioavailability.
Any statin might be treated in such a way, however, the oral bioavailability of pravastatin and rosuvastatin (which are hydrophilic) appears to be less likely to benefit from such treatment. The lipophilic drugs cerivastatin and pravastatin have per se a high bioavailability (of approximately 60%) which could be enhanced by micro crystallization.
The statin will normally be administered orally or by any parenteral route, in the form of a pharmaceutical formulation comprising the active ingredient, optionally in the form of a non-toxic organic, or inorganic, acid, or base, addition salt, in a pharmaceutically acceptable dosage form. Depending upon the disorder and patient to be treated, as well as the route of administration, the medicament may be administered at varying doses.
hi human therapy, the statin can be administered alone but will generally be administered in admixture with a suitable pharmaceutical excipient diluent or carrier selected with regard to the intended route of administration and standard pharmaceutical practice.
For example, the statin can be administered orally, buccally or sublingually in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavouring or colouring agents, for immediate-, delayed- or controlled- release applications. The stain may also be administered via intracavernosal injection.
Such tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycollate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropyhnethylcellulose (HPMC), hydroxy-propylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included.
Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, a cellulose, milk sugar or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the statin may be combined with various sweetening or flavouring agents, colouring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
The statin can also be administered parenterally, for example, intravenously, intra- arterially, intraperitoneally, intrathecally, intraventricularly, intrasternally, intracranially, intra-muscularly or subcutaneously, or they may be administered by infusion techniques. They are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well-known to those skilled in the art.
Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
The statin can also be administered intranasally or by inhalation and are conveniently delivered in the form of a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray or nebuliser with the use of a suitable propellant., e.g. dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoro-ethane, a hydrofluoroalkane such as 1,1,1,2-tetrafluoroethane (HFA 134A3 or 1,1,1,2,3,3,3-heptafluoroρropane (HFA 227EA3), carbon dioxide or other suitable gas. In the case of a pressurised aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. The pressurised container, pump, spray or nebuliser may contain a solution or suspension of the active compound, e.g. using a mixture of ethanol and the propellant as the solvent, which may additionally contain a lubricant, e.g. sorbitan trioleate. Capsules and cartridges (made, for example, from gelatin) for use in an inhaler or insufflator may be formulated to contain a powder mix of a statin and a suitable powder base such as lactose or starch.
Aerosol or dry powder formulations are preferably arranged so that each metered dose or "puff contains precise quantity of statin for delivery to the patient. It will be appreciated that he overall daily dose with an aerosol will vary from patient to patient, and may be administered in a single dose or, more usually, in divided- doses throughout the day.
Alternatively, the statin can be administered in the form of a suppository or pessary, or they may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder. The statin may also be transdermally administered, for example, by the use of a skin patch. They may also be administered by the ocular route, particularly for treating diseases of the eye.
For ophthalmic use, the statin can be formulated as micronised suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzylalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.
For application topically to the skin, the statin can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene polyoxypropylene compound, emulsifying wax and water. Alternatively, they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavoured basis, usually sucrose and acacia or tragacanth; pastilles comprising the active ingredient in an inert basis such as gelatin and glycerin, or sucrose and acacia," and mouth-washes comprising the active ingredient in a suitable liquid carrier.
Generally, in humans, oral or topical administration of the statin is the preferred route, being the most convenient. In circumstances where the recipient suffers from a swallowing disorder or from impairment of drug absorption after oral administration, the drug may be administered parenterally, e.g. sublingually or buccally.
For oral and parenteral administration to human patients, the daily dosage level of statin are provided above.
The physician in any event will determine the actual dosage which will be most suitable for any individual patient and it will vary with the age, weight and response of the particular patient. The above dosages are exemplary of the average case. There can, of course, be individual instances where higher or lower dosage ranges are merited and such are within the scope of this invention.
As mentioned above, the medicament can be used for the prevention or treatment of cachexia. Existing treatment for this disorder include the following therapeutic agents: growth hormone, anabolic steroids, and appetite stimulants for some subgroups of cachexia.
In a further embodiment of the invention the medicament comprises one or more of such therapeutic agents for preventing or treating cachexia.
A further aspect of the invention provide a method of treating a patient with or at risk of developing cachexia comprising administering an appropriate quantity of at least one statin, wherein the patient is human or a domestic or farm animal. Preferably the patient is human.
An embodiment of this aspect of the invention is wherein the patient is administered an amount of statin that does not substantially reduce cholesterol level.
An embodiment of this aspect of the invention is wherein the statin is atorvastatin, fmvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, cerivastatin, mevastatin or pitavastatin. Preferably the statin is simvastatin or atorvastatin. An embodiment of this aspect of the invention is wherein the method uses a quantity of the statin equivalent to a daily unit dose.
An embodiment of this aspect of the invention is wherein the method uses a slow release formulation of the statin having the property of releasing a daily unit dose.
An embodiment of this aspect of the invention is wherein the statin has been prepared using micro-crystallisation.
An embodiment of this aspect of the invention is wherein the method further comprises using one or more further therapeutic agents for preventing or treating cachexia.
An embodiment of this aspect of the invention is wherein the cachexia is cancer cachexia, cardiac cachexia or chronic obstructive pulmonary disease (COPD) cachexia.
The medicament or method is considered to be useful in assisting weight gain. Example 2 shows a higher likelihood of experiencing weight gain (assessed as weight gain of more than 5%) for patients treated with statins. Example 3 also shows a positive trend for low dose statin on body weight.
The medicament or method is considered to be useful in assisting increase in lean tissue or skeletal muscle mass. Example 3 shows a positive trend for low dose statin on lean tissue and skeletal muscle mass.
An example (NMR scans) of how lean tissue mass can be measured is described in Example 3.
Muscle mass can be measured in humans in several ways, as will be well known to the person skilled in the art, for example: - antropometry (using calipers to assess skin fold thickness and then estimate muscle mass)
- DEXA scanning (dual X-ray absorptiometry, as described in, for example Anker et al (1999) 20, 683-693). DEXA is the method most often used in cachexia research projects
- CT scanning (for example as described in Anker et al (1997) Eur Heart J 18(2), 259-69)
- MRI scanning.
The skeletal muscle mass increase may be relative to the starting point of muscle mass in the patient or as the proportion of the patient's body mass. From a muscle mass and strength point of view either is desirable. Preferably after treatment there is more muscle than before: whether this is accompanied by an increase in body weight or not is of lesser importance for muscle anabolic interventions. It is preferable that muscle increases without loss of fat, i.e. that muscle gain leads to overall weight gain.
AU documents referred to herein are hereby incorporated by reference.
The invention will now be described by reference to the following, non-limiting Examples and Figures.
Figure 1. Chemical structures of various statins. Mevastatin is not in clinical use, but it is largely used in in vitro studies. Pravastatin is currently approved in Japan only.
Figure 2: Pathway of cholesterol biosynthesis. Statins block the rate-limiting step in the cascade by inhibition of HMG-CoA reductase. The inhibition can be overcome by addition of mevalonate in experimental models. CoA - coenzyme A, HMG- 3-hydroxy-3-methylglutaryl, PP - pyrophosphate. Figure 3: Overview of the principal pleiotropic effects of statins on (A) the endothelium, (B) the immune system, and (C) tumour development and metastasis. Several statins have been shown to improve endothelial function by increasing nitric oxide (NO) production and release. Statins also increase the number of endothelial progenitor cells in the bloodstream. Production of proinflammatory mediators (e.g. TNF-a, IL-I, IL-6) and adhesion molecule expression (e.g. ICAM-I) are all reduced with statin treatment in experimental models. The release of C-reactive protein (CRP) has been proven in several clinical settings. Anti-tumour activities of statins comprise the induction of apoptosis, growth arrest by inhibiting the cell cycle of malignant cells, and the reduction of the invasiveness of malignant cells at secondary sites.
Example 1: Characteristics of statins
The following table includes information on the characteristics of statins that may be used in the invention.
TABLEl:
Figure imgf000020_0001
O
Figure imgf000021_0001
to
O
Figure imgf000022_0001
Figure imgf000023_0001
* The 80 mg dose of fluvastatin is available as an extended release formulation only.
** Cmax may vary according to the administered dose and the patient population investigated. to
Example 2: Use of statins for the treatment of cachexia
In an analysis of the ELITE 2 trial database (Pitt et al., (2000) Lancet 355, 1582- 1587), we found that in patients with heart failure who also have COPD statins are associated with a higher likelihood of experiencing weight gain >5% independently of tables below.
The statins used in the ELITE 2 trial were:
- atorvastatin , n=79, 10-40 mg/d (10 unknown)
- cerivastatin , n=12 , 0.2-0.3 mg/d (1 unknown)
- fluva , n=43 , 20-80 mg/d (6 unknown)
- prava , n=141, 1-80 mg/d (16 unknown)
- simva , n=300 , 5-80 mg/d (27 unknown) - lova, n=51 , 10-60 mg/d (3 unknown)
This data is presented in the following tables.
RESULTS: ELITE 2 study. Cox Proportional Hazard Analysis of the impact of being on statin at baseline (BL) on the subsequent development of weight gain >5%. The impact is highly significant (p<0.0001) and independent of the severity of heart failure as measured by LVEF, NYHA class, clinical oedema status, degree of kidney dysfunction (i.e. creatinine [crea] levels) and heart failure aetiology. In 3030 patients all information for this analysis was available.
Table 2
Figure imgf000024_0001
Figure imgf000025_0001
Model Coefficients for FU days gain 5%. Censor Variable: gain 5% y=0/no=l. Model: Proportional Hazards. Row exclusion: ELITE2 ll-05B-w-change-080805.
Below is a survival summary table for Table 2.
Figure imgf000025_0002
Censor Variable: gain 5% y=0/no=l. Model: Proportional Hazards. Row exclusion: ELITE2 11- 05B-w-change-080805.
Below are the individual hazard ratios and their 95% confidence interval related to the analysis in Table 2
Figure imgf000025_0003
Confidence Intervals for FU days gain 5%. Censor Variable: gain 5% y=0/no=l. Model: Proportional Hazards. Row exclusion: ELITE2 l l-05B-w-change-080805.
RESULTS AND CONCLUSIONS: ELITE 2 study - the subgroup of patients with a diagnosis of chronic obstructive pulmonary disease (COPD).
Cox Proportional Hazard Analysis of the impact of being on statin at baseline on the subsequent development of weight gain >5%. The analysis below in Table 3 shows an important trend for a 33.4% increase in the occurrence of >5% weight- gain when a patient was on a statin.
These results are independent of the severity of heart failure as measured by LVEF, NYHA class, clinical oedema status, and the degree of kidney dysfunction (i.e. creatinine [crea] levels). This analysis was performed on 259 patients with COPD and in 60 patients a weight gain >5% event occurred.
Importantly this analysis shows a) that good cardiac function (e.g. high LVEF) was not related to experiencing weight gain. In fact per % increase in LVEF a 2.1% decrease in the frequency of >5% weight gain was observed, and
B) that statin therapy was associated with weight gain only in patients with COPD (33.4% increase in the event rate of weight gain), unlike the general population of the ELITE 2 trial in which the occurrence of weight gain due to statin therapy was 17% reduced (see Table 2).
Table 3
Figure imgf000026_0001
Model coefficients for FU days gain 5%. Censor Variable: gain 5% y=0/no=l . Model: Proportional Hazards. Row exclusion: ELITE2 ll-05B-w-ch-COPD-090805.
Below are the individual hazard ratios and their 95% confidence interval related to the analysis in
Table 3
Figure imgf000026_0002
Confidence Intervals for FU days gain 5%. Censor Variable: gain 5% y=0/no=l. Model: Proportional Hazards. Row exclusion: ELITE2 ll-05B-w-ch-COPD-090805. Example 3: Effect of Simvastatin in Cancer Cachexia
1) Material and methods Animal model The ascites hepatoma Yoshida AH-130 cells (108) were inoculated into 200 g male Wistar rats. The animals are housed in groups of three. The day after inoculation animals are randomized into various groups. The rats then receive treatment with simvastatin in 4 doses: O.lmg, l.Omg, 10 mg and 20 mg vs placebo. The primary endpoints of the study include assessment of body weight, body composition and survival. Organ weight is assessed at the end of the study (or after death) as a secondary endpoint. The animals were killed 14 days after tumour inoculation.
Time line
Day -2/-l Locomotor activity
Day 0: NMR (Body composition), Echocardiography
Day 1 : Tumor-inoculation
Day 2: Randomisation, start therapy
Day 3 : NMR (Body composition) Day 6: NMR (Body composition)
Day 9: NMR (Body composition)
Day 10/11 : Locomotor activity
Day 12: NMR (Body composition), Echocardiography
Day 15: NMR (Body composition), end of therapy, sacrifice, storage of tissue and plasma
Analysis, primary endpoints
Survival
The animals were seen at least twice per day to ensure precise survival data.
Body Weight Body weight was monitored every two days throughout the entire study period.
Analysis, secondary endpoints
Body Composition Body composition was measured the day before tumour inoculation and in two day intervals after that until the end of the study using NMR-scans (EchoMRI systems). Read-out is fat, lean mass, total water and free water. Locomotor Activity Assessed using Supermex system over a period of 24 hours in cages where the animals are placed individually (type 3 cage).
Heart function / echocardiography
Echocardiography was performed in all animals on day 0 and on day 12 using a high resolution echocardiography system (Vevo770, Visual Sonics) to determine the following parameters:
LVEF (left ventricular ejection fraction)
LVFS (left ventricular fractional shortening) LVEDD and LVESD (left ventricular end-diasolic and end-systolic diameters) septum and inferior wall thickness (M-Mode) rVSd and IVSs (thickness of the inter- ventricular septum in diastole and systole) diastolic filling (E-wave)
Stroke Volume Organ weights
The weight of the heart, skeletal muscle (gastrocnemius, soleus, EDL, tibialis) and liver was assessed whether at the end of the study or post-mortem.
2) Results
Survival
Neutral results were observed in the survival analysis. Very few events occurred during the 14-day study period indicating that the model may have been shorter than optimal and no statistical test could be applied.
Events: l Plac l of lO
O.lmg l of l l l.Omg O of 11 lOmg 1 of 15
20mg O of 14
Heart function Heart function was assessed by high resolution echocardiography using the Vevo 770 system (Visualsonics, Toronto, Canada). The left ventricular (LV) ejection fraction, LV fractional shortening, LV end-diastolic diameter, LV end-systolic diameter, LV posterior wall thickness in diastole and systole, thickness of the inter-ventricular septum in diastole and systole, the LV stroke volume and the diastolic function (E/ A) were assessed. The results showed neutral results for all parameter assessed (ANOVA, all p>0.4).
Body weight
The ANOVA did not show a significant difference between the treatment groups. However, a trend was observed in body weight (without tumour) for O.lmg simvastatin (mean + 8.1% vs placebo; p=0.23)
Fat and lean tissue mass (NMR assessment)
Fat mass did not show any significant difference between the simvastatin groups and the placebo group. The ANOVA for lean mass was also not significant, but a trend was seen for the O.lmg simvastatin group (mean +8.2% vs placebo, p=0.26).
Organ weight
While the weights of the liver, brown fat as well as white fat did not shown any differences, the skeletal muscle mass of important leg muscles of the O.lmg simvastatin group showed a positive trend vs placebo (Musculus gastrocnemius mean +16%; ρ=0.18; Musculus tibialis mean +10.7%; ρ=0.083).
Tumour size (total cell number) The cell number showed a trend in the ANOVA analysis (p=0.18) and the cell number was lower in the l.Omg simvastatin group vs placebo (mean -19.3%; ρ=0.091). Table 4: Results - parameters other than heart function
Figure imgf000030_0001
Table 5: heart function results
Figure imgf000031_0001
3) Summary In summary, only the low dose simvastatin showed positive trends, while the high ■ • dose has detrimental effects on the animals in this cancer cachexia model.

Claims

Claims
1. The use of at least one statin in the manufacture of a medicament for the prevention or treatment of cachexia in a patient, wherein the patient is human or a domestic or farm animal.
2. The use of claim 1 wherein the patient is human.
3. The use of claim 1 or 2 wherein the medicament is for administering an amount of statin that does not substantially reduce cholesterol level.
4. The use of any of the previous claims wherein the statin is atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin, simvastatin, cerivastatin, mevastatin or pravastatin.
5. The use of claim 4 wherein the statin is simvastatin or atorvastatin.
6. The use of any of the previous claims wherein the medicament comprises a quantity of the statin equivalent to a daily unit dose.
7. The use of any of the previous claims wherein the medicament a slow release formulation of the statin having the property of releasing a daily unit dose.
8. The use of any of the previous claims wherein the statin has been prepared using micro-crystallisation.
9. The use of any one of the previous claims wherein the medicament further comprises one or more further therapeutic agents for preventing or treating cachexia.
10. The use of any one of the previous claims wherein the cachexia is cancer cachexia, cardiac cachexia (for example cachexia due to CHF or cardiomyopathy), chronic obstructive pulmonary disease (COPD) cachexia, or cachexia associated with liver cirrhosis, chronic renal failure, rheumatoid arthritis, diabetes, stroke, burns, trauma or other acute injury, sepsis, HIV/ AIDS or other infections or ageing.
11. The use of any one of the preceding claims wherein the medicament is for treating patients selected as above 60, 65, 70 or 15 years, of age.
12. A method of treating a patient with or at risk of developing cachexia comprising administering an appropriate quantity of at least one statin, wherein the patient is human or a domestic or farm animal.
13. The use of any one of claims 1 to 11 or method of claim 12 wherein the medicament or method is for assisting weight gain.
14. The use of any one of claims 1 to 11 or 13 or method of claim 12 or 13 wherein the medicament or method is for assisting increase in lean tissue or skeletal muscle mass.
PCT/GB2007/000146 2006-01-18 2007-01-18 Methods WO2007083119A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2008550841A JP2009523778A (en) 2006-01-18 2007-01-18 Method
AU2007206776A AU2007206776A1 (en) 2006-01-18 2007-01-18 Methods
EP07704928A EP1973542A2 (en) 2006-01-18 2007-01-18 Treatment of cachexia with statins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0600967.4A GB0600967D0 (en) 2006-01-18 2006-01-18 Methods
GB0600967.4 2006-01-18

Publications (2)

Publication Number Publication Date
WO2007083119A2 true WO2007083119A2 (en) 2007-07-26
WO2007083119A3 WO2007083119A3 (en) 2008-03-20

Family

ID=36010486

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2007/000146 WO2007083119A2 (en) 2006-01-18 2007-01-18 Methods

Country Status (5)

Country Link
EP (1) EP1973542A2 (en)
JP (1) JP2009523778A (en)
AU (1) AU2007206776A1 (en)
GB (1) GB0600967D0 (en)
WO (1) WO2007083119A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009121623A2 (en) * 2008-04-04 2009-10-08 Summit Corporation Plc Compounds for treating muscular dystrophy
EP2832357A1 (en) * 2013-07-29 2015-02-04 Charité - Universitätsmedizin Berlin Selective AT2 receptor agonists for use in treatment of cachexia
US9486503B2 (en) 2012-10-04 2016-11-08 Shionogi & Co., Ltd. Medicinal agent for suppressing malignant tumor metastasis
US10675274B2 (en) 2018-09-19 2020-06-09 Forma Therapeutics, Inc. Activating pyruvate kinase R
US10836771B2 (en) 2017-03-20 2020-11-17 Forma Therapeutics, Inc. Compositions for activating pyruvate kinase
US11001588B2 (en) 2018-09-19 2021-05-11 Forma Therapeutics, Inc. Activating pyruvate kinase R and mutants thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003007993A1 (en) * 2001-07-19 2003-01-30 Pharmacia Corporation Combination of an aldosterone receptor antagonist and an hmg coa reductase inhibitor

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003007993A1 (en) * 2001-07-19 2003-01-30 Pharmacia Corporation Combination of an aldosterone receptor antagonist and an hmg coa reductase inhibitor

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ANKER STEFAN D ET AL: "Statin use and survival in patients with chronic heart failure--results from two observational studies with 5200 patients." INTERNATIONAL JOURNAL OF CARDIOLOGY 20 SEP 2006, vol. 112, no. 2, 20 September 2006 (2006-09-20), pages 234-242, XP002464984 ISSN: 1874-1754 *
DAVIS M P ET AL: "Cachexia and anorexia: cancer's covert killer." SUPPORTIVE CARE IN CANCER : OFFICIAL JOURNAL OF THE MULTINATIONAL ASSOCIATION OF SUPPORTIVE CARE IN CANCER MAY 2000, vol. 8, no. 3, May 2000 (2000-05), pages 180-187, XP002464985 ISSN: 0941-4355 *
MAHMOUD F A ET AL: "The role of C-reactive protein as a prognostic indicator in advanced cancer" CURRENT ONCOLOGY REPORTS, CURRENT SCIENCE, GB, vol. 4, no. 3, May 2002 (2002-05), pages 250-255, XP008087414 ISSN: 1523-3790 cited in the application *
MUSCARITOLI MAURIZIO ET AL: "Effects of simvastatin administration in an experimental model of cancer cachexia." NUTRITION (BURBANK, LOS ANGELES COUNTY, CALIF.) 2003 NOV-DEC, vol. 19, no. 11-12, November 2003 (2003-11), pages 936-939, XP002464981 ISSN: 0899-9007 cited in the application *
PITT B ET AL: "Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure: randomised trial--the Losartan Heart Failure Survival Study ELITE II." LANCET 6 MAY 2000, vol. 355, no. 9215, 6 May 2000 (2000-05-06), pages 1582-1587, XP002464986 ISSN: 0140-6736 cited in the application *
PITT B ET AL: "Effects of losartan versus captopril on mortality in patients with symptomatic heart failure: rationale, design, and baseline characteristics of patients in the Losartan Heart Failure Survival Study--ELITE II." JOURNAL OF CARDIAC FAILURE JUN 1999, vol. 5, no. 2, June 1999 (1999-06), pages 146-154, XP002464987 ISSN: 1071-9164 *
RIDKER P M ET AL: "Long-term effects of pravastatin on plasma concentration of C-reactive protein. The Cholesterol and Recurrent Events (CARE) Investigators." CIRCULATION 20 JUL 1999, vol. 100, no. 3, 20 July 1999 (1999-07-20), pages 230-235, XP002464983 ISSN: 1524-4539 *
SASAKI MAKOTO ET AL: "The 3-hydroxy-3-methylglutaryl-CoA reductase inhibitor pravastatin reduces disease activity and inflammation in dextran-sulfate induced colitis." THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS APR 2003, vol. 305, no. 1, April 2003 (2003-04), pages 78-85, XP002464982 ISSN: 0022-3565 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009121623A2 (en) * 2008-04-04 2009-10-08 Summit Corporation Plc Compounds for treating muscular dystrophy
WO2009121623A3 (en) * 2008-04-04 2010-05-20 Biomarin Iga Limited Compounds for treating muscular dystrophy
US9486503B2 (en) 2012-10-04 2016-11-08 Shionogi & Co., Ltd. Medicinal agent for suppressing malignant tumor metastasis
US9987333B2 (en) 2012-10-04 2018-06-05 Shionogi & Co., Ltd. Method for suppressing malignant tumor metastasis
EP2832357A1 (en) * 2013-07-29 2015-02-04 Charité - Universitätsmedizin Berlin Selective AT2 receptor agonists for use in treatment of cachexia
WO2015014634A1 (en) * 2013-07-29 2015-02-05 Charite - Universitätsmedizin Berlin Selective at2 receptor agonists for use in treatment of cachexia
US10016397B2 (en) 2013-07-29 2018-07-10 Charité—Universitaetsmedizn Berlin Selective AT2 receptor agonists for use in treatment of cachexia
US10836771B2 (en) 2017-03-20 2020-11-17 Forma Therapeutics, Inc. Compositions for activating pyruvate kinase
US11014927B2 (en) 2017-03-20 2021-05-25 Forma Therapeutics, Inc. Pyrrolopyrrole compositions as pyruvate kinase (PKR) activators
US11396513B2 (en) 2017-03-20 2022-07-26 Forma Therapeutics, Inc. Compositions for activating pyruvate kinase
US11649242B2 (en) 2017-03-20 2023-05-16 Forma Therapeutics, Inc. Pyrrolopyrrole compositions as pyruvate kinase (PKR) activators
US10675274B2 (en) 2018-09-19 2020-06-09 Forma Therapeutics, Inc. Activating pyruvate kinase R
US11001588B2 (en) 2018-09-19 2021-05-11 Forma Therapeutics, Inc. Activating pyruvate kinase R and mutants thereof
US11071725B2 (en) 2018-09-19 2021-07-27 Forma Therapeutics, Inc. Activating pyruvate kinase R
US11844787B2 (en) 2018-09-19 2023-12-19 Novo Nordisk Health Care Ag Activating pyruvate kinase R
US11980611B2 (en) 2018-09-19 2024-05-14 Novo Nordisk Health Care Ag Treating sickle cell disease with a pyruvate kinase R activating compound

Also Published As

Publication number Publication date
GB0600967D0 (en) 2006-03-01
JP2009523778A (en) 2009-06-25
AU2007206776A1 (en) 2007-07-26
EP1973542A2 (en) 2008-10-01
WO2007083119A3 (en) 2008-03-20

Similar Documents

Publication Publication Date Title
JP7445893B2 (en) C. for the treatment of solid tumors in humans. novyi
EP1973542A2 (en) Treatment of cachexia with statins
JP2001514224A (en) Combination therapy including amlodipine and statin compound
JP2016528275A (en) Composition and therapy for promoting plaque regression
JP2010500283A (en) Methods of treating inflammatory diseases using tyrosine kinase inhibitors
JP2009511639A (en) Composition for lowering serum cholesterol and / or serum triglycerides
US20070149578A1 (en) Combination Therapy
US8048880B2 (en) Treatment of cardiovascular disease and dyslipidemia using secretory phospholipase A2 (SPLA2) inhibitors and SPLA2 inhibitor combination therapies
US20050261246A1 (en) Methods for treating inflammatory and autoimmune diseases
US6844361B2 (en) Pharmaceutical composition comprising a sodium hydrogen exchange inhibitor and an angiotensin converting enzyme inhibitor
JP2021102624A (en) Treatment of cyr61- and vegf-mediated conditions
TW200306853A (en) Therapeutic agent for glomerular disease
US8399518B2 (en) Administration of 3,5-diiodothyropropionic acid for stimulating weight loss, and/or lowering triglyceride levels, and/or treatment of metabolic syndrome
US20100204249A1 (en) Secretory phospholipase a2 (spla2) inhibitor and niacin drug compositions and methods for treating cardiovascular disease and dyslipidemia
US20050182036A1 (en) Medicinal composition containing an HMG-CoA reductase inhibitor
US8383612B2 (en) Use of megestrol acetate for improving heart function and the treatment of heart insufficiency
US20110136835A1 (en) Combination formulations of tranilast and allopurinol and methods related thereto
US9345671B2 (en) Adiponectin production enhancer
JP2010526152A (en) Treatment of cardiovascular disease and dyslipidemia using secretory phospholipase A2 (SPLA2) inhibitor and SPLA2 inhibitor combination therapy
WO2004096276A9 (en) Sugar intake-ability enhancer
JPWO2014034871A1 (en) Preventive or therapeutic agent for dyslipidemia
JP4607436B2 (en) Pharmaceutical composition containing an HMG-CoA reductase inhibitor
US20240148735A1 (en) Methods of treating heart failure with vibegron
KR20020091245A (en) Antihypertensive Agents and Use
US20150104416A1 (en) Leukemic Stem Cell Ablation

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2008550841

Country of ref document: JP

Ref document number: 2007704928

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007206776

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2007206776

Country of ref document: AU

Date of ref document: 20070118

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2007206776

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2007704928

Country of ref document: EP