ZA200105838B - Drug combinations comprising (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino] pyrimidin-5-YL] (3R,5S) -3,5-dihydroxyhept-6-enoic acid and an inhibitor inducer or substrate of P450 isoenzyme 3A4. - Google Patents

Drug combinations comprising (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino] pyrimidin-5-YL] (3R,5S) -3,5-dihydroxyhept-6-enoic acid and an inhibitor inducer or substrate of P450 isoenzyme 3A4. Download PDF

Info

Publication number
ZA200105838B
ZA200105838B ZA200105838A ZA200105838A ZA200105838B ZA 200105838 B ZA200105838 B ZA 200105838B ZA 200105838 A ZA200105838 A ZA 200105838A ZA 200105838 A ZA200105838 A ZA 200105838A ZA 200105838 B ZA200105838 B ZA 200105838B
Authority
ZA
South Africa
Prior art keywords
drug
isoenzyme
inhibitor
substance
combination
Prior art date
Application number
ZA200105838A
Inventor
Ali Raza
Howard Gerard Hutchinson
Takahiko Baba
Yoshitaka Yamaguchi
John Stuart Pears
Dennis Schneck
Akira Touchi
Original Assignee
Astrazeneca Ab
Shionogi & Co
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 Astrazeneca Ab, Shionogi & Co filed Critical Astrazeneca Ab
Publication of ZA200105838B publication Critical patent/ZA200105838B/en

Links

Landscapes

  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Description

DRUG COMBINATIONS COMPRISING (E)-7-[4-(4-FLUOROPHENYL )-6-ISOPROPYL~2-[METHYL(
METHYLSULFONYL )AMINOJPYRIMIDIN-5-YL](3R,5S)-3,5-DIHYDROXYHEPT-6-ENOIC ACID
AND AN INHIBITOR, INDUCER OR SUBSTRATE OF P450 ISOENZYME 3A4
The invention concerns safe non-interacting drug combinations of a 3-hydroxy-3- methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, which is (E)-7-[4- (4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino]pyrimidin-5-y1] (3R,5S)-3,5- dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof (the Agent) and a drug which is either an inducer, inhibitor or a substrate of cytochrome P450, in particular cytochrome P450 isoenzyme 3A4. Particular combinations are useful in treating hyperlipidaemia in humans who are receiving immunosuppressive chemotherapy. A preferred combination is the Agent and a fibrate drug, the use of such a combination in treating hyperlipidaemia in mammals, and medicaments containing such a combination for use in such treatments.
Hypercholesterolaemia is one of the strongest risk factors for atherosclerosis which is ’ 15 associated with coronary artery disease (including angina pectoris, myocardial infarction and mortality), stroke (including cerebro vascular accident and transient ischaemic attack) and : peripheral arterial occlusive disease. Several types of hypercholesterolaemia exist. The magnitude of hypercholesterolaemia may have consequences for the therapy, but in general, any reduction of elevated plasma cholesterol levels is generally accepted to result in an improvement of the risk profile. Dietary improvement and increased exercise are essential first steps and should continue even if drug therapy is instituted, but the therapeutic potential of drug therapy is significantly larger. Several types of drug therapy for hypercholesterolaemia are currently available. Guidelines exist for the treatment of hypercholesterolaemia for example, American Heart Association (AHA) (Anon 1988),
Updated Sheffield treatment tables (Heart (1998) 80 Supp.2 S1-S29) and Recommendations of the task force of the European Society of Cardiology Guidelines (Pyorala 1994).
HMG-CoA reductase inhibitors are the most widely used prescription medication for the treatment of hypercholesterolaemia. By inhibiting the rate-controlling step in cholesterol biosynthesis, these agents effectively lower the plasma concentrations of atherogenic particles containing cholesterol such as low-density lipoprotein (LDL-C) and very low-density
-2- oo lipoprotein (VLDL-C). Partial inhibition of hepatic cholesterol synthesis causes up-regulation of hepatic membrane LDL-C receptors which are responsible for the clcarance of LDL-C from the circulation. In addition, reduced hepatic synthesis of cholesterol is thought to result in a modest reduction in the secretion of VLDL-C particles by the liver. Clinical trials with certain HMG Co A-reductase inhibitors, such as in the Scandinavian Simvastatin Survival
Study, confirm a reduction in cardiovascular morbidity and mortality with such agents, and may even promote regression of atherosclerotic vascular lesions. Various HMG Co A- reductase inhibitors arc marketed, and are collectively referred to as ‘statins’.
Despite the impressive benefits of statin therapy, less than optimal therapeutic results may be achieved in some subjects, particularly in the more severe classes of hypercholesterolaemia.
This can be due to the occurrence of reversible increases in liver transaminase levels at higher dose levels of statins as well as differences in efficacy between different statins. Clinically important (>3 times upper limit of normal [ULN]) elevations in serum alanine aminotransferase [ALT]) have been reported for atorvastatin in 0.8 per cent of patients at low ) doses of atorvastatin and higher at raised doses (European Summary of Product
Characteristics [SmPC] for atorvastatin [Lipitor™]). In all cases the effect is dose-related and ) reversible. In general it is the incidence of ALT increases which limits dose escalation of statins rather than a limit to further increases in efficacy.
The first generation statins (such as lovastatin, pravastatin and simvastatin - prodrug derivatives of fungal metabolites - and fluvastatin) are categorised in that they achieve only a limited cholesterol lowering affect before the dose administered is limited by elevations in serum ALT. Second generation “superstatins” (such as atorvastatin - synthetic compounds- structurally distinct from first generation compounds) inhibitors are categorised in that they lower cholesterol levels to a much higher degree than the earlier first generation of statins before their dose is limited by serum ALT levels. Atorvastatin has been successful over the first generation of statins. Since its launch in the USA atorvastatin has reached sales in 1998, doubling from 1997, of $2.2 billion, capturing 38% of new prescriptions for cholesterol- lowering agents in the US and is now the most widely prescribed hypolipidaemic agent in the
US (Wamner-Lambert 1998 annual results).
An additional adverse event, reported for statins in general, is myopathy, defined as symptoms of muscle pain, tenderness and weakness, with creatinine kinase (CK) values >10 x Upper
Limit of Normal (ULN). This adverse event is not considered to be dose related, and in addition the adverse events are potentially more serious, and consequently more problematical. In severe cases this can lead to thabdomyolysis, which is a rare life threatening condition sometimes associated with renal failure. The incidence of raised CK levels >10x
ULN - on 2 occasions at least 1 week apart with symptoms = myositis according to FDA) for statins has been reported as 3.1 per cent. (SmPC for atorvastatin). Myopathy and rhabdomyolysis have been particularly associated with taking a statin in combination with gemfibrozil, niacin, cyclosporin or erythromycin, (Hunninghake H. Et al. Current Opinion in
Lipidolgy (1992), 3, 22-28) which are all substrates for P450 isoenzyme 3A4. The increase in adverse events associated with taking a combination of a statin drug with one of the other drugs mentioned above is probably due to a drug:drug interaction likely related to the : 15 metabolism of most statins also by the same cytochrome P450 isoenzyme 3A4. Therefore when a drug which is also metabolised by P450 3A4 is administered alongside a statin which - also 1s metabolised by P450 3A4, the side effects discussed above are more likely to occur.
Increase in the side effects, such as muscle damage, is thought to be due to elevated statin levels in muscle cells inhibiting farnesylation and geranylgeranylation of muscle proteins .
Elevated levels of statins may be caused by any drug which affects P450 3 A4. Therefore, currently on the labels of all commercially available statins the use of the statin in combination with drugs that are metabolised by P450 3A4 is not recommended and is contraindicated in certain cases.
Nearly all drugs are metabolised to some degree in the human, generally to a less lipid soluble compound which is more easily excreted by the kidney or in liver bile. The liver is the major site of drug metabolism and many drug metabolising enzymes occur at high concentration in the endoplasmic reticulum (which form microsomes upon homogenisation) of liver parenchymal cells (hepatocytes). Cytochrome P450 represents a major class of drug metabolising enzymes and exists as a family of isoenzymes found in hepatic microsomes. Six specific P450 isoenzymes are responsible for the metabolism of most of the commonly used
-4- Co drugs, namely P450 1A2, 2C9, 2C19, 2D6, 2E1 and 3A4.
A major disadvantage of the currently available “super statin” , atorvastatin, is that atorvastatin is metabolised by cytochrome P450 enzymes, in particular 3A4, which may cause drug interactions with other drugs which are inducers, inhibitors or substrates of the same P450 enzyme which metabolises atorvastatin. All of the first generation of statins are metabolised by P450 also. However, the rate of metabolism of pravastatin is sufficiently low that it is considered less susceptible to clinically relevant drug interactions. Therefore despite the lower efficacy of pravastatin, in its currently available doses, at reducing hypercholesterolaemia this is currently the statin of choice in combination with other drugs : where the possibility of drug interactions is unacceptably high. (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino}pyrimidin-5-y1] (3R,58)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof (the calcium salt of which is disclosed in Figure 1 below), hereinafter referred to as the Agent, is ) also a statin and belongs to the class of what is now starting to be called a “superstatin”.
The Agent is disclosed in European Patent Application, Publication No. 0521471, and in
Bioorganic and Medicinal Chemistry, (1997), 5(2), 437-444 as an inhibitor of HMG-CoA reductase which is a major rate-limiting enzyme in cholesterol biosynthesis. The Agent is described as useful in the treatment of hypercholesterolaemia, hyperlipoproteinaemia and atherosclerosis.
The Agent is not metabolised by cytochrome P450 3A4 and therefore does not possess the same potential for drug interaction shared with the currently available “super statin”, i.e. atorvastatin, or any of the other currently available statins.
Therefore we present as a feature of the invention a non-interacting drug combination comprising a HMG CoA reductase inhibitor, which is the Agent, and a drug which is an inhibitor, inducer or substrate of P450 in particular, isoenzyme 3A4.
. Cs.
As a further feature of the invention we present use of a HMG CoA reductase inhibitor, which is the Agent, in the preparation of a medicament for use in combination therapy with a drug which is an inhibitor, inducer or substrate of P450, in particular, isoenzyme 3A4.
As a further feature of the invention we present use of a drug which is an inhibitor, inducer or substrate of P450, in particular, isoenzyme 3A4 in the preparation of a medicament for use in combination therapy with a HMG CoA reductase inhibitor, which is the Agent.
As a further feature of the invention we present a pharmaceutical formulation comprising the
Agent, a drug which is an inducer, inhibitor or substrate of P450 isoenzyme 3A4 and a pharmaceutically-acceptable diluent, carrier or adjuvant.
As a further feature of the invention we present a pharmacy pack comprising a first drug which is the Agent and a second drug which is an inducer, inhibitor or substrate of P450 ) 15 isoenzyme 4A4. ; By the term “inducer of P450 ” we mean a drug which increases the rate at which a P450 enzyme, in particular isoenzyme 3A4, metabolises a substrate, for example by increasing the activity of the P450 enzyme, decreasing the rate of biological inactivation of the P450 enzyme or by increasing the rate of transcription of the P450 gene.
By the term “inhibitor of P450” we mean a drug which lowers the rate at which a P450 enzyme, in particular isoenzyme 3A4, metabolises a substrate, for example by lowering the activity of the P450 enzyme or by lowering the rate of transcription of the P450 gene.
By the term “substrate of P450”” we mean a drug which is metabolised by a P450 enzyme, in particular isoenzyme 3A4.
By the term “non-interacting drug combination” we mean a drug combination for which there is no adverse affect to the patient by its administration through the mechanism of drug metabolism by cytochrome P450 isoenzyme 3A4. It is recognised that in certain instances a drug interaction may nevertheless occur between two such drugs when in combination
B through a completely different mechanism not involving drug metabolism, such as affecting drug absorption.
Whether a drug is an inhibitor, inducer or substrate of a P450 enzyme can be easily determined by procedures known to the skilled person. Such procedures may involve the exposure of a radiolabelled drug to hepatocytes or hepatocyte microsomes or isolated P450 enzyme and the use of analytic techniques, such as HPLC, in determining metabolite formation. A specific procedure is described herein. . By the term “combination” we mean either that the Agent and the drug of the combination : are administered together in the same pharmaceutical formulation or that the Agent and the : drug are administered separately. When administered separately components of the 3 combination may be administered to the patient simultaneously or sequentially. - We have found that the Agent is not metabolised significantly by the major cytochrome P450 isoenzymes 1A2, 2C9, 2C19, 2D6 and 3A4. This is a further feature of the invention. .
Preferred non-interacting combinations of the invention include those in which the Agent is combined with a drug which is also involved in lowering cholesterol and is also an inducer, inhibitor or substrate of P450 3A4. Examples include fibrates, such as bezafibrate, clofibrate, ciprofibrate, fenofibrate and gemfibrizol (preferably fenofibrate), and niacin. Specific embodiments of this preferred feature are described in Section B below.
Preferred non-interacting combinations of the invention include those in which the Agent is combined with a drug which is involved in treating cardiovascular conditions and which is also an inhibitor, inducer or substrate of P450 3A4. Examples include digitoxin, diltiazem, losartan, nifedipine, quinidine, verapamil and warfarin.
Preferred non-interacting combinations of the invention include those in which the Agent is combined with cyclosporin and /or tacrolimus (FK506) and therefore has utility in treating
. Ly elevated cholesterol levels in patients who are about to, or have recently undergone, a transplantation operation. Specific embodiments of this preferred feature are described below.
Preferred patients in which the combination of the invention is to be administered are those who suffer from myopathy or rhabdomylosis or who have already been found to suffer from myopathy or rhabdomylosis when treated with HMG Co A reductase inhibitor which is metabolised by P450 3A4, for example atorvastatin, simvastatin and lovastatin.
Other features of the invention include those described above wherein the Agent is used at doses of 5 to 80mg per day. When a dose range of 5 to 80 mg per day is referred to herein for the Agent other particular dosage ranges, which are further independent aspects of the invention, include (as appropriate) 10 to 80 mg per day, 10 to 60 mg per day, 10 to 40 mg per day, 5 to 40 mg per day, 5 to 20 mg per day, 10 to 20 mg per day, 20 to 60 mg per day, 20 to 40 mg per day and 40 to 60 mg per day. Particular dosages are 5, 10, 20, 40 and 80mg per : 15 day. A particularly suitable starting dose of the Agent in the methods referred herein is 5 to 10 mg per day, especially 10 mg per day.
P450 3A4 substrates include; acetominophen, aldrin, aflentanil, amiodorane, astemizole, benzphetamine, budenoside, carbamazepine, cyclophosphamide, cyclosporin, dapsone, digitoxin, ditiazem, diazepam, erthromycin, etoposide, flutamide, hydroxyarginine, ifosphamide, imipramine, lansoprazole, lidocaine, lovatidine, losartan, lovastatin, midrazolam, nifedipine, omeprazole, quinidine, rapamycin, retenoic acid, steroids, tacrolimus, teniposide, theophyline, toremifene, triazolam, troleandomycin, verapamil, warfarin, zatosetron and zonisamide.
P450 3A4 inhibitors include; clotrimazole, ethinylestradiol, gestodene, itraconazole, ketoconazole, miconazole, diltiazem, naringenin, erthromycin, cyclosporin and triacetyloleandomycin.
P450 3A4 inducers include; carbamazepine, dexamethasone, phenobarbital, phenytoin, rifampin, sulfadimidine, sulfinipyrazone and triacetyloleandomycin.
Examples of other P450 inducers, inhibitors or substrates include those mentioned in
Drug Metabolism Reviews (1997) Vol 29, Issue 1+2, pages 413-580, Rendic, S. and Di Carlo,
F.J. “Human cytochrome P450 enzymes,: A status report summarising their reactions, substrates, inducers and inhibitors”.
Dosages of the Agent may be administered according to the cholesterol lowering effect desired from a range of 5 to 80 mg per day in any number of unit dosages. Dosages of the - drug which is an inducer, inhibitor or substrate of P450 3A4 are those which are advised for : each drug, or which are commercially available. Advantageously, due to the lack of interaction at the level of P450 3A4, the skilled person may dose the Agent with a drug which . is an inducer, inhibitor or substrate of P450 3A4 with out needing to make any adjustments. ) a The dose ranges and dosages described above are further independent features of the invention. ) - Preferably the Agent is bis[(E)-7-[4-(4-fluorophenyl)-6-isopropyl-2- [methyl(methylsulfonyl)amino]pyrimidin-5-y1J(3R,5S)-3,5-dihydroxyhept-6-enoic acid] calcium salt (illustrated in figure 1).
Experimental
The experiment below is used to determine the in vitro metabolic fate of [*C]- labelled Agent in human hepatocytes and, in addition, to determine the specific P450 isozymes involved in [“C]- labelled Agent metabolism, if any. The latter experiment involves an investigation of the effects of P450 selective chemical inhibitors (see Table 1) on the metabolism of [“C]- labelled Agent.
. 0. :
COMPOUND: ['*C]- labelled Agent.
Chemical name: Bis [(E)-7-[4-(4-fluorophenyl)-6-isopropyl-2- [methyl(methylsulfonyl)amino]pyrimidin-5-y1J(3R,5S)-3,5- dihydroxyhept-6-enoic acid] calcium salt
Isomer: 3R,58,6F Stereoisomer
Molecular weight: 1001.16 (Ca salt)
Formulation ingredients: The labelled Agent is dissolved in water to produce a solution suitable for addition to the incubates.
TISSUE SOURCE Human liver, suitable for the preparation of microsomes and hepatocytes, obtained from The International Institute for the
Advancement of Medicine (Exton, USA). Human hepatocytes may, in addition, be obtained from Biowhittaker Ltd. or United
Kingdom Human Tissue Bank (Leicester, England). ) 15
EXPERIMENTAL PROCEDURES
(1) METABOLISM OF ['“C]- LABELLED AGENT BY HUMAN HEPATOCYTES ["“C)- labelled Agent (1 pM or higher concentration if required for analytical sensitivity) was incubated with hepatocytes in culture obtained from two human organ donors. Cultures were terminated with ethanol after 0, 6, 24 and 48 hours of incubation and stored at approximately -20°C until analysed. The metabolic competence of the hepatocytes was confirmed at the time of incubation by examining their ability to metabolise ['*C]-ethoxycoumarin (25 uM); aliquots were removed into methanol at the same time points as for the test compound.
Following incubation of ['“C]-ZD4522 with hepatocytes, metabolite profiles were generated by High Performance Liquid Chromatography (HPLC). The ability of hepatocytes to metabolise ['*C]-ethoxycoumarin was confirmed by HPLC.
: ASSESSMENT OF DATA on Data generated was assessed with regard to the following :- (1) Assess whether human hepatocytes metabolise ['“C]- labelled Agent. (2) Quantitate the amount of each metabolite formed. (2) ENZYMES INVOLVED IN METABOLISM OF THE AGENT [“C]- labelled Agent (at an appropriate concentration) was incubated with human hepatic microsomes in the absence and presence of selective P450 inhibitors (see Table 1). Similar incubations of ['“C]- labelled Agent with individual heterologously expressed P450 isoenzymes was also performed. Incubations were terminated by the addition of an - 10 appropriate organic solvent. Metabolite profiles of the incubates are generated by HPLC.
Table 1 Selective chemical inhibitors of P450 isozymes = P450 isozyme Selective inhibitor }
En 1A2 Furafylline 2C9 Sulphaphenazole 2C19 Omeprazole 2D6 Quinidine 3A4 Ketoconazole
ASSESSMENT OF DATA
Data generated during this study was assessed with regard to the following:- (a) Therate and extent of metabolism of [“C)- labelled Agent. (b) The ability of the selective P450 inhibitors to decrease the metabolism of [“C]- labelled Agent was compared in order to determine the isozyme(s) involved in the metabolism of ['*C]- labelled Agent.
The ability of individual expressed P450 isoforms to metabolise ['*C]- labelled Agent was assessed to aid determination of the P450 isozyme(s) involved in the metabolism of ['*C]- labelled Agent. (c) These in vitro data can be used to predict the variability of the pharmacokinetics of the
Agent in the population and the likely effects on the pharmacokinetics of the Agent during co-administration with known P450 enzyme inhibitors/inducers.
It was found that the Agent was not significantly metabolised by whole hepatocytes and that this was inhibited by sulphaphenazole and omeprazole.
FOR TREATING HYPERLIPIDAEMIA AND ASSOCIATED CONDITIONS IN POST TRANSPLANT
PATIENTS RECEIVING IMMUNOSUPPRESSIVE THERAPY.
Two common drugs used in suppressing the human immune system, cyclosporin and . 15 tacrolimus (formerly called FK506), are known to be metabolised by cytochrome P450 3A4.
In particular cyclosporin is also a known inhibitor of P450 3A4 and is therefore likely to } reduce the metabolism of any other drug which is metabolised by P450 3A4.
Therefore where immunosuppressive therapy is prescribed, such as with the drugs cyclosporin and tacrolimus (especially cyclosporin), the attendant physician must be cautious as to any other therapy which may be jointly presented to the patient in combination.
Immunosuppressive therapy is most commonly used before, during and after human transplant operations. In particular with cardiac transplants the attendant physician may wish to also place the patient on statin drug therapy to reduce future incidents of coronary heart disease, stroke, peripheral arterial occlusive disease or peripheral vascular disease, particularly in patients with elevated cholesterol or in normolipidaemic patients with other risk factors associated with heart disease. In particular within this special patient group (human transplant patients), the patients arc at high risk of developing accelerated atherosclerosis in the transplant organ in an aggressive fashion and within a short period of time due, in part, to the surgical damage to the blood vessels during transplantation, any previously underlying untreated conditions and the immunosupressive therapy. Hyperlipidaemia is common after transplantation even in patients who did not suffer hyperlipidaemia prior to transplantation, incidence 60-80% of recipients. a
-12- oo
It is known that certain immunosuppresive drugs, such as steroids, cyclosporin and tacrolimus, raise cholesterol levels in patients (Wierzbicki AS (1999) IICP 53 (1) 54-59). In addition cyclosporin and tacrolimus may raise the levels of fibrinogen and lipoprotein (a) in 3 the patient, further accelerating the progression of atherosclerosis in the transplant patient (Hohaye H, Clin.Transplant (1997) 11, 225-230 and Hilbrands LB, J .Am.Soc.Nephroi (1995) + 5,2073-2081). This unusually accelerated atherosclerosis is present in about 20% of heart transplant patients at 1 year and 40-65% at 5 years (Chang G. Et al. American Heart Journal (1998), 136(2), 329-334). The incidence of accelerated atherosclerosis has been reported as causing a 1-18% incidence of CHD at one year and 20-50% incidence of CHD at 3 years in cardiac transplant patients (Erdoes LS, J.Vasc.Surg. (1995) 22, 434-440). Lovastatin, = pravastatin and simvastatin have all shown to lower cholesterol levels in heart transplant . patients. In a placebo controlled study pravastatin increased survival of transplant paticnts by 1 year and significantly reduced the incidence of haemodynamic organ rejection. Because of - the lower incidence of serious drug interaction with the immunosuppresive therapy pravastatin - 15 is currently the statin drug of choice in post transplant treatment regimes. However, as ) i discussed above, pravastatin does not lower lipid/cholesterol levels to such a great extent as, for example, atorvastatin. :
We have discovered that the Agent is extremely effective at treating hypercholesterolacmia in patients following transplantation and that the Agent is not metabolised by cytochrome P450 isoenzyme 3A4. Therefore we have found through the use of the Agent in a clinical study that the Agent may be conveniently dosed to patients who are undertaking immunosuppressive therapy without any clinically significant side effects associated with the concomitant dosing of the Agent and the immunosuppressive drug(s) and, in addition, also achieve much higher levels of cholesterol lowering than has previously been achieved, such as by the use of pravastatin,
We present as the first feature of the invention a method of providing safe non-interacting cholesterol lowering therapy to a human patient undertaking immunosuppressive chemotherapy which method comprises administering to the patient the Agent.
Particular patients undertaking immunosuppressive chemotherapy who may benefit from the method of the invention are those who: 1) suffer primary (type IIa) hypercholesterolaemia (LDL-L > 135 and TG<200); 2) suffer combined (type IIb) hypercholesterolaemia (LDL-C> 135 and TG>200); 3) patients with established CHD or other atherosclerotic disease, such a PVD, stroke or penpheral arterial occlusive disease; 4) patients who are at high risk of developing CHD or other atherosclerotic disease, such as described above, because of a combination of risk factors. The term “high risk” is defined in the “Recommendations of Second Joint Task Force of European and other Societies on Coronary Prevention”, (Wood, D. et. al. European Heart ’ 15 Journal, Atherosclerosis and Journal of Hypertension 1998) as absolute CHD risk of 2 20% over 10 years or will exceed 20% if projected to age 60 years. Whether a : patient is at high risk or not may be determined by the charts which accompany the above recommendations and which charts are incorporated herein by reference. For example a male patient in his 40s who smokes and has a systolic blood pressure of 180 mm Hg or higher and a total plasma cholesterol concentration of 7 mmol/L or higher will be classified as high risk. Similarly other guidelines for reducing risk factors may be applied such as those described in; a) JAMA, June 16, 1993-Vol 629, No.23, Pages 3015-3023 - “Summary of the
NCEP Adult Treatment Panel II Report” - specifically Figure 1. Page 3018-3019 which is incorporated herein by reference. b) Post Graduate Medical Journal 1993; 69(811): 359-369 - “Management of hyperlipidaemia: guidelines of the British Hyperlipidaemic Association”- specifically
Table V and Table VI which are incorporated herein by reference.
c) Heart 1998; 80 Supplement 2:S1-S29 - “Joint British recommendations on prevention of coronary heart disease in clinical practice” - specifically Figure 1 on = pages S4-S5, which is incorporated herein by reference. oC d) The Lancet 1995; December 2, Vol.346, 1467-1471 - “Sheffield risk and or 5 treatment table for cholesterol lowering for primary prevention of coronary heart - disease” - specifically the Table appearing at page 1468, which is incorporated herein by reference. 5) patients who suffer type I or II diabetes; - 6) patients who are about to or have already undertaken a heart transplant;
The statin therapy may be administered so as to achieve in the patient undertaking oC immunosuppressive chemotherapy.
BST or 1) A reduction in the internal thickness of coronary artery atheroma of > 30% as measured by IVUS. 2) A reduction of LDL-C of at least 30, 40, 50%. 3) A maintenance or increase of HDL-C of at least 5, 10, 15%. 4) A change in any of the above values better than pravastatin at a similar dose and over the same period.
As a further feature of the invention, and due also to the fact that the Agent is not metabolised to any significant extent by P450 isoenzymes, it is possible to administer, more safely than before, to a patient receiving immunosuppresive therapy a fibrate and the Agent. As discussed earlier the administration of a fibrate and a statin has previously been associated with a higher incidence of rhabdomyolysis and myopathy. In addition fibrate drugs do interact with cyclosporin due to both being metabolised by the same P450 isoenzyme. Therefore, the use of a statin and a fibrate drug in combination with immunosuppresive therapy was previously ~ contraindicated due to the likelihood of possible serious interactions (Hunninghake 1992,
Wanner C. Kidney Int. (1995) 52(suppl.), 860-S62; and Katznelson S. Contributions Nephrol. (1997) 120, 97-104). However, if possible, it would be advantageous to also administer a fibrate alongside a statin since fibrates are known to lower different lipoproteins than statins and therefore their combined pharmacology would be complementary in reducing even further the likelihood of CHD and other diseases mentioned above associated with the formation of atherosclerosis. Therefore the possibility of combining the Agent, which is not metabolised by
P450 3A4, with a fibrate and an immunosuppresive therapy offers the additional possibility of lowering cholesterol to a greater extent in such patients than previously achieved and more safely than could previously be achieved by the administration of a statin, a fibrate and an immunosuppresive drug.
Fibrate drugs are thought to act through peroxisomal proliferating activator receptor-a ] 15 (PPAR-a) and affect gene activation at a number of genes involved in atheroma. Patients on fibrate drugs show improved LDL subfraction distribution (reduced VLDL and raised HDL), } reduced LDL and reduced triglyceride levels, and possible advantages through improving insulin sensitivity. Examples of fibrate drugs include, bezafibrate, ciprofibrate, fenofibrate and gemfibrozol.
By use of the term “safe non-interacting statin therapy” we mean that the Agent is not metabolised by P450 3A4 and therefore does not affect the metabolism of the immunosuppresive therapy or vice versa.
Diseases and conditions in which immunosuppressive therapy may be prescribed include, in addition to organ transplantation mentioned above, autoimmune diseases, including rheumatic disorders, such as, rheumatoid arthritis, osteoarthritis, lupus erthematosus; and other autoimmune disorders such as idiopathic thrombocytopenic purpura, autoimmune haemolytic anaemia and acute glomerulonephritis.
Er
The agent may be administered at the same time as the immunosuppressive chemotherapy, or if not at the same time within a short time period of administration of the immunosuppressive therapy, such as in the same day, within 6, 3, 2 or 1 hour.
The Agent may be administered according to the cholesterol lowering effect desired from a range of 5-80 mg per day in any number of unit dosages, preferable once a day dosing. Ideal doses are 10, 20 and 40 mg once per day. Preferred doses are 20 and 40mg once per day.
Particular immunosuppressive drugs which may be combined with the Agents are those which are metabolised by liver enzymes, such as by P450 3A4, and therefore are not likely to have a drug interaction with the Agent. Examples include those described above, cyclosporin and tacrolimus, as well as corticosteroids, which are also metabolised in the liver. Examples of corticosteroids include prednisone (especially used for organ transplantation). Preferably at least one of the immunosuppressive agents, if more than one agent is used, is either cyclosporin or tacrolimus, preferably cyclosporin. )
Example
The following non-limiting example is of a clinical trial to demonstrate the performance of this aspect of the invention.
PROTOCOL
Title: A Double-blind, Parallel Group Study to Assess the
Change in Coronary Artery Atheroma Burden Post
Cardiac Transplantation as Measured via IVUS after 12
Months Dosing with the Agent versus Pravastatin
Objectives: The primary objective of the study is to measure change in maximal mean intimal thickness of the anterior descending coronary artery as assessed by intravascular ultrasonography (IVUS) (read centrally) after 12 months of treatment with the Agent or pravastatin. A change from baseline of >30% in intimal thickness is considered clinically significant.
The secondary objectives of the study are to measure the effects on coronary artery atheroma burden and to compare effects of the Agent with the following assessments: ® cvidence of organ rejection as assessed by adverse event reports. » measurement of LDL-C, HDL-C, apoB, apoA-I, Lp (a) concentrations, ex vivo platelet aggregation, fibrinogen, PAI-I, and the concentrations of circulating markers of vascular inflammation. e comparison of lipid values after 52 weeks of treatment. e measurement of inflammatory markers after 52 weeks of treatment (HLA antigen VCAM/ICAM expression as assessed by biopsy). e to determine the drug’s safety and tolerability.
Type and number of subjects: Approximately 40 men and women (aged 18 years and older) post cardiac transplant with hypercholesterolemia and triglycerides <400 mg/dl at the time of randomisation.
Trial treatment: Once daily doses of the Agent (10 mg) or pravastatin (10 mg) for two weeks, then titration of dose to 20 mg of the Agent or pravastatin 20 mg. After 4 weeks the dose should be titrated up to 40 mg of the Agent or 40 mg pravastatin. Patients who have had their dose titrated up to 40 mg may have their dose titrated down to 20 mg, at the discretion of the investigator.
Duration of treatment: Eligible subjects randomised to 1 of 2 treatment groups, the Agent or pravastatin, for 52 weeks.
: -18- oo
Primary measure: Mean change from baseline in maximal mean intimal thickness, as assessed by IVUS (read centrally).
Secondary measures: Percent change from baseline in LDL-C at 6 and 12 months.
Percent change from baseline in total cholesterol (TO), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), LDL-C/HDL-C,
TC/HDL-C, non-HDL-C/HDL-C, and triglycerides (TG).
Percent change from baseline in ApoB, ApoB/ApoA-1,
ApoA-1, Lp (a), and particle subfractions at 6 and 12 months.
Percentage of subjects on each of the possible titrated doses at 12 months. }
Endocardial rejection will be considered an adverse event.
Percent change from baseline in inflammatory markers (HLA antigen level and ICAM/VCAM expression).
Safety evaluation as determined by adverse events, physical examination, and laboratory data.
TRIAL DESIGN
This is a multicenter, randomized, double-blind, parallel-group clinical trial. Within 1 to 4 weeks post surgery, subjects are randomized to receive either the Agent or pravastatin for 52 weeks. Subjects start treatment at a dose of 10 mg of either the Agent or pravastatin at
Visit 2 and the dose is titrated to 20 mg at Visit 3 during the forced titration period. At Visit 4 and subsequent visits, the investigator has the option to increase each drug up to 40 mg during the optional titration period. Patients who have had their dose titrated up to 40 mg may have their dose titrated back down to 20 mg at the investigator’s discretion.
TRIAL DESIGN
Pre- Forced Optional Titration transplant Titration
Visit 1 2 3 4 5 6 7 8 9 10 11
Week (W)/ Wo wW2 wW4 M2 M3 M4 M5 M6 M9 M12
Month (M)
Agent (mg) 10 20 =20*
PRAVASTATIN (mg) 10 20 220%
Randomisation** * Subjects who are tolerating 20 mg of the Agent or Pravastatin at Visit 4 may have their dose titrated up to 40 mg, at the discretion of the investigator. ** Subjects should be randomized within 4 weeks of cardiac transplantation and must not have received any other lipid lowering therapy post-surgery.
Inclusion criteria 5) have undergone cardiac transplantation up to four weeks prior to randomization (2) fasting TG concentrations of <4.52 mmol/L (400 mg/dl)
Exclusion criteria
Any of the following is regarded as a criterion for exclusion from the trial;
D Use of other cholesterol lowering drugs or lipid lowering dietary supplements or food additives post-transplantation prior to entering the study 2) history of serious or hypersensitivity reactions to other HMG-CoA reductase inhibitors (3) pregnant women, women who are breast feeding, and women of child bearing potential who are not using chemical or mechanical contraception or have positive serum pregnancy test (a serum -Human chorionic gonadotropin [B-HCG] analysis) id
0) Subjects with a history of diabetic ketoacidosis within the past 5 years are excluded. ®) uncontrolled hypothyroidism defined as a thyroid stimulating hormone (TSH) >1.5 times the ULN at Visit 2 or subjects whose thyroid replacement therapy was initiated within the last three months (6) use of concomitant medications as detailed below - except immune suppressants and diazepam
UE current alcohol and/or drug abuse ®) active liver disease or hepatic dysfunction as defined by elevations of > 1.5 times the
ULN at Visit 2 in any of the following liver function tests: ALT, AST, or bilirubin (9) serum CK > 3 times ULN at Visit 2 (10) serum creatinine > 220 pmol/L (2.5 mg/dl) (11) subjects with cancer or with a history of cancer who, in the opinion of the investigator, have more than a minimal chance of recurrence (12) participation in another investigational drug trial less than 4 weeks before randomization into the trial (13) subjects randomized to double-blind treatment who subsequently withdrew cannot re-enter this trial (14) serious or unstable medical or psychological conditions that, in the opinion of the investigator, would compromise the subject’s safety or successful participation in the trial (15) subjects taking cyclic hormone replacement therapy (HRT), cyclic oral contraceptive therapy (OCT), a depot progesterone injection, or subjects whose non-cyclic HRT or
OCT was initiated within the last 3 months
DISALLOWED MEDICATIONS
Antibiotics/ antifungals Erythromycin Base
Erythromycin Ethyl Succinate, Acetyl
Sulfisoxazole
Rifampicin
Fluconazole
Ketaconazole
Itraconzole
Anti-epileptics/ antidepressants Phenytoin
Phenobarbitol
Fluoxetine
Carbemazepine
Antiulcer drugs Cimetidine ——
El i —
Systemic Steroids Triamcinolone Acetonide
Triamcinolone Diacetate
Betamethasone
Sodium Phosphate
Betamethasone Acetate
Hydrocortisone
Hydrocortisone Acetate
Hydrocortisone Sodium Phosphate
Hydrocortisone Sodium Succinate
Cortisone Acetate
Dexamethasone
Dexamethasone Acetate
Dexamethasone Sodium
Prednisone
Methylprednisolone
Methylprenisolone Acetate
Methylprednisolone Sodium
Succinate - Prednisolone Tebutate
Prednisolone Sodium Phosphate
Methyltestosterone
Fluoxymesterone
Antihistamine Astemizole i
CLASS OF DRUG GENERIC NAME
Lipid Regulation Niacin/Nicotinic Acid
Probucol
Psyllium Preparations
Clofibrate
Cholestyramine
Colestipol Hydrochloride
Gemfibrozil
Atorvastatin
Lovastatin
Pravastatin (except study medication)
Simvastatin
Fluvastatin
Cerevestatin : Fish oils (any dose) lipid lowering dietary supplements . lipid lowering food additives
Hormone Therapy Estrogen and progesterone combinations which are bi or tri phasic.
Friedewald Equation
The LDL-C level is calculated from the Friedewald equation as follows:
For SI units (mmol/l)
LDL-C = Total cholesterol - [HDL-C + Triglycerides/2.2)
For non-SI units (mg/dl):
LDL-C = Total cholesterol - [HDL-C + triglycerides/5]
-24- oC
Summary of NCEP Goals for Lipid Management®
NCEP Risk Category Target LDL-C (NCEP)
No CHD/PVD and 1 or no risk factors < 160 mg/dL
No CHD/PVD and 2 or more risk factors < 130 mg/dL
Clinically evident CHD/PVD < 100 mg/dL * Second Report of the Expert Panel on Detection, Evaluation, and Treatment of High Blood . Cholesterol in Adults. Bethesda (MD): National Institutes of Health, National Heart and Lung
Institute 1993 Sep Report No.: 93-3095.
NCEP National Cholesterol Education Program. }
FOR TREATING HYPERLIPIDAEMIA, AND ASSOCIATED CONDITIONS, USING A COMBINATION
OF THE AGENT AND A FIBRATE DRUG OR NIACIN
Myopathy and rhabdomyolysis have been associated with taking a statin in combination with gemfibrozil, niacin, cyclosporin or erythromycin, (HMG CoA reductase inhibitors,
Hunninghake, Current Opinion in Lipidology (1992) 3, 22-28) which are all substrates for
P450 3A4. Additionally, adverse events associated with taking a fibrate drug have also been reported to increase with concomitant statin therapy, such as a myosistis-flu like syndrome, which occasionally occurs in patients receiving gemfibrozil, increases to 5% of patients when a statin is also administered.
Combination of a statin with a fibrate drug is contraindicated on the labels, both in the USA and Europe, of all commercially available statins. Despite the possibility of the occurrence of serious drug interactions doctors do prescribe combination therapy of a statin and a fibrate drug to patients with more severe levels of hypercholesterolaemia, such as in patients with familial combined hyperlipidaemia, where the risk of a serious drug interaction is outwei ghed by the benefits of the combination therapy. It is recommended that where combination therapy of a fibrate drug and a statin is prescribed that patients should have their CK value determined on a regular basis, typically every 6-weeks, until a stable pattern is established. Therapy is stopped if muscle symptoms occur in association with elevated CK activity. However, as quoted from the US label of Lipitor™ “there is no assurance that such monitoring [of CK levels] will prevent the occurrence of severe myopathy”.
We have discovered that the Agent is extremely effective at treating mixed hyperlipidaemia and hypertriglyceridaemia in patients when combined with a fibrate drug and that the Agent is not metabolised by cytochrome P450 isoenzyme 3A4. Therefore we have found through the use of the Agent in a clinical study that the Agent may be conveniently dosed to patients who are also taking a fibrate drug without any clinically significant side effects associated with the concomitant dosing of the Agent and the fibrate drug. In addition much higher levels of lipid - 15 lowering than has previously been achieved can be achieved by the use of the Agent and a fibrate drug. The combination is of most use in mixed hyperlipidemia where the LDL and . VLDL and TGs are all elevated.
We present as the first feature of the invention a method of providing safe non-interacting lipid lowering combination therapy to a mammal, including a human patient, preferably a patient suffering mixed hyperlipidaemia and hypertriglyceridaemia, which method comprises administering to the patient the Agent and a fibrate drug or niacin.
By the term “combination” as used herein we mean either (1) that the Agent and the fibrate drug of the combination are administered together in the same pharmaceutical formulation or (2) that the Agent and the drug are administered separately. When administered separately components of the combination may be administered to the patient simultaneously or sequentially.
By the term “fibrate drug” we mean the class of drugs which are based around the structure/activity of fibric acid and such drugs include the following commercially available versions; bezafibrate, clofibrate, ciprofibrate, fenofibrate and gemfibrizol, preferably fenofibrate.
Preferred patients in which the combination of the invention is to be administered arc those who have already been found to suffer from myopathy or rhabdomylosis when treated with a statin and/or with a fibrate drug which is metabolised by P450 3A4.
Particular patients who may benefit from the method of the invention are those who: 1) suffer combined (type IIb) hypercholesterolaemia (typically LDL-C> 135 mg/dL and
TG2200 mg/dL); 2) suffer familial (type IV and V) hypercholesterolaemia; 3) patients suffering secondary hypercholesterolaemia from such conditions as: a) diabetes (type I or II), b) nephrotic syndrome, : c) uremia, d) hyperthyroidism, and : €) obstructive liver disease. 4) patients with established CHD or other atherosclerotic disease, such a PVD, stroke or peripheral arterial occlusive disease; 5) patients who are at high risk of developing CHD or other atherosclerotic disease, such as described above, because of a combination of risk factors. The term “high risk” is defined in the “Recommendations of Second Joint Task Force of European and other Societies on Coronary Prevention”, (Wood, D. et. al. European Heart Journal, Atherosclerosis and
Journal of Hypertension 1998) as absolute CHD risk of > 20% over 10 years or will exceed 20% if projected to age 60 years. Whether a patient is at high risk or not may be determined by the charts which accompany the above recommendations and which charts are incorporated herein by reference. For example a male patient in his 40s who smokes and has a systolic blood pressure of 180 mm Hg or higher and a total plasma cholesterol concentration of 7 mmol/L or higher will be classified as high risk. Similarly other guidelines for reducing risk factors may be applied such as those described in;
a) JAMA, June 16, 1993-Vol 629, No.23, Pages 3015-3023 - “Summary of the
NCEP Adult Treatment Panel II Report” - specifically Figure 1. Page 3018-3019, which is incorporated herein by reference. b) Post Graduate Medical Journal 1993; 69(811): 359-369 - “Management of hyperlipidaemia: guidelines of the British Hyperlipidaemic Association” specifically
Table V and Table VI, which are incorporated herein by reference. c) Heart 1998; 80 Supplement 2:51-S29 - “Joint British recommendations on prevention of coronary heart disease in clinical practice” - specifically Figure 1 on pages 84-55, which is incorporated herein by reference. d) The Lancet 1995; December 2, Vo0l.346, 1467-1471 - “Sheffield risk and treatment table for cholesterol lowering for primary prevention of coronary heart disease” - specifically the Table appearing at page 1468, which is incorporated herein by reference. : 15 The statin therapy may be administered so as to achieve in the patient receiving a fibrate drug or niacin: 1) a reduction of LDL-C of at least 30, 40, 50, 60, 70 or 80%. 2) a maintenance or increase of HDL-C of at least 5, 10, 15%. 3) a reduction in triglycerides of at least 10, 20, 30 or 40%.
The combination of the fibrate, or niacin, and the Agent may be applied as separate dosage forms, which may be taken simultaneously or sequentially, or in a combined dosage form.
The combination of the fibrate and the Agent will also have an additive or synergistic effect on the reduction in LDL-C, maintenance or increase of HDL-C or reduction in tri glyceride in the patients blood.
In addition the combination of niacin and the Agent may be applied as separate dosage forms, which may be taken simultaneously or sequentially, or in a combined dosage form. The combination of the fibrate and the Agent will also have an additive or synergistic effect on the reduction in LDL-C, maintenance or increase of HDL-C or reduction in triglyceride in the patients blood.
Doses of the Agent which are administered are at the discretion of the attendant physician generally taking into account the severity of the disease, the age, weight and sex of the patient.
However typical doses will be from 5 to 80 mg per day orally, preferably as a once a day oral tablet form.
Doses of the fibrate drug or niacin which are administered in the combination of the invention also are at the discretion of the attendant physician taking into account all of the above factors plus in particular which drug is used.
For clofibrate (such as Atromid-S®) 20-30 mg/kg body weight daily in 2 or 3 divided oral doses after meals is typical.
For bezofibrate (such as Bezalip®) 400 mg once a day orally, after food at night or in the morning, is typical. )
For fenofibrate (such as Lipantil®) 200 mg once a day, or 62 mg three times a day, with food is typical.
For gemfibrozil (such as Lopid®) 600 mg twice a day orally is typical.
For cipofibrate (such as Modalim®) 100 mg once a day orally is typical.
For niacin (NIASPAN®, an extended release niacin formulation, and preferred feature) 500mg once to four times daily, preferably twice or four times daily.
A preferred fibrate drug is fenofibrate.
Preferably the AGENT is administered to a patient receining niacin at 10mg or 40mg daily doses.
The particular aspect of this invention is illustrated by the following non-limiting examples:
Clinical Trial
A Randomised, Non-controlled, Single-centre, Open-label, 3-way Crossover Trial to Assess the
Effect of Co-administration of the Agent and Fenofibrate on the Pharmacokinetics of Each
Compound in Healthy Male Volunteers
Objectives: The primary objective of this trial is to assess the effect of co- administration of the Agent and fenofibrate on the pharmacokinetics of both the Agent and fenofibrate
The safety of all volunteers will be ensured by clinical monitoring
Type and number of 14 healthy male volunteers volunteers:
Trial design: The trial will be a randomised, non-controlled, 3-way crossover study . carried out at a single centre
Trial treatment: This trial will consist of three 7-day treatment periods (Periods A, B, and C). Volunteers will receive, in random order, a 10 mg capsule of the Agent once daily for 7 days, a 67 mg fenofibrate capsule 3 times daily for 7 days and the combination for 7 days.
There will be a minimum of a 3-week washout between each trial period.
Duration of The study will consist of 3 periods of 7-day dosing (a total of 21 treatment: dosing days) with a 3-week washout between dosing in Periods A, B and C.
Primary endpoints: The primary endpoints are: e AUC(0-24) and C,,, of the Agent in the presence and absence of fenofibrate ¢ AUC(0-8) and C,,, of fenofibrate in the presence and absence of the Agent
Secondary the secondary endpoints are: endpoints: ® tan tos Coy, for the Agent in the presence and absence of ) fenofibrate ® tae tins Cy fOr fenofibrate in the presence and absence of the
Agent * safety assessments: symptoms, blood pressure and pulse rate, ECG, clinical chemistry, haematology and urinalysis
TRIAL PLAN
Summary of procedures - overall plan for Trial Periods A, B and C
Trial Days | Medical | Doses of the P&BP | 12lead | Safety Blood | Kinetics of | Kinetics
Agent / ECG & Urine the Agent | Fenofibrate fenofibrate or combination
Il I i I
I EE I I I
I I I
I EE I I I I A
IE RN I I I
I I I I
I I I I I A
I I I I I
I EE I I I I
I EE I I I
I I I I
I I I sl I I I I °Full clinical chemistry, haematology and urine labstix. "Clinical chemistry only: urea, creatinine, total protein, albumin, uric acid, total bilirubin (and unconjugated and conjugated bilirubin if total bilirubin raised), alkaline phosphatase, alanine aminotransferase (ALT), aspartate aminotranseferase (AST), gamma glutamyltransferase, creatine kinase (CK), sodium, potassium, calcium, cholesterol and triglycerides. “Pre-dose all trial periods. “Only trial periods when volunteers receive the Agent eonly trial periods when volunteers receive fenofibrate
P = pulse; BP = blood pressure a

Claims (47)

  1. Claims
    I. A non-interacting drug combination comprising a HMG-CoA reductase inhibitor, which is (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino]pyrimidin-5- y1] (3R,58)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof and a drug which is an inhibitor, inducer or substrate of P450 isoenzyme 3A4.
  2. 2. A non-interacting drug combination, as claimed in claim 1, wherein the second drug is an inhibitor or inducer of P450 isoenzyme 3A4.
  3. 3. A non-interacting drug combination, as claimed in either claim 1 or claim 2, wherein each drug is administered together or each drug is administered sequentially.
  4. 4. A non-interacting drug combination, as claimed in any claim from 1 to 3, wherein the : 15 second drug is used to lower cholesterol and is an inducer, inhibitor or substrate of P450 isoenzyme 3A4.
  5. 5. A non-interacting drug combination, as claimed in claim 4, wherein the second drug is selected from bezafibrate, clofibrate, fenofibrate, gemfibrozol and niacin.
  6. 6. A non-interacting drug combination, as claimed in claim 5, wherein the second drug is fenofibrate.
  7. 7. A non-interacting drug combination, as claimed in any claim from 1 to 3, wherein the second drug is used in treating cardiovascular conditions and is also an inhibitor, inducer or substrate of P450 isoenzyme 3A4.
  8. 8. A non-interacting drug combination, as claimed in claim 7, wherein the second drug is selected from digitoxin, diltiazem, losartan, nifedipine, quinidine, verapamil and warfarin.
  9. 9. A non-interacting drug combination, as claimed in any claim from 1 to 3, wherein the second drug is used in immunosuppresion therapy and is an inducer, inhibitor or substrate of P450 isoenzyme 3A4. 5
  10. 10. A non-interacting drug combination, as claimed in claim 9, wherein the second drug - is selected from cyclosporin, tacrolimus and a corticosteroid.
  11. 11. A non-interacting drug combination, as claimed in any claim from 1 to 10, wherein (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino]pyrimidin-5-yl] (3R,5S)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof is dosed + at 5, 10, 20, 40 or 80mg once per day.
  12. 12. A pharmaceutical formulation comprising (E)-7-[4-(4-fluoropheny1)-6-isopropyl-2- i [methyl(methylsulfonyl)amino}pyrimidin-5-y1] (3R,5 S)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof, a drug which is an inducer, inhibitor or substrate of P450 isoenzyme 3A4 and a pharmaceutically-acceptable diluent, carrier or adjuvant.
  13. 13. A pharmaceutical formulation, as claimed in claim 12, wherein the second drug is a substrate of P450 isoenzyme 3A4 and is selected from acetominophen, aldrin, aflentanil, amiodorane, astemizole, benzphetamine, budenoside, carbamazepine, cyclophosphamide, cyclosporin, dapsone, digitoxin, ditiazem, diazepam, erthromycin, etoposide, flutamide, hydroxyarginine, ifosphamide, imipramine, lansoprazole, lidocaine, lovatidine, losartan, lovastatin, midrazolam, nifedipine, omeprazole, quinidine, rapamycin, retenoic acid, steroids, tacrolimus, teniposide, theophyline, toremifene, triazolam, troleandomycin, verapamil, warfarin, zatosetron and zonisamide.
  14. 14. A pharmaceutical formulation, as claimed in claim 12, wherein the second drug is an inhibitor of P450 isoenzyme 3A4 and is selected from clotrimazole, ethinylestradiol, gestodene, itraconazole, ketoconazole, miconazole, diltiazem, naringenin, erthromycin, cyclosporin and triacetyloleandomycin.
  15. 15. A pharmaceutical formulation, as claimed in claim 12, wherein the second drug is an inducer of P450 isoenzyme 3A4 is selected carbamazepine, dexamethasone, phenobarbital, phenytoin, rifampin, sulfadimidine, sulfinipyrazone and triacetyloleandomycin.
  16. 16. A pharmacy pack comprising a first drug which is (E)-7-[4-(4-fluorophenyl)-6- isopropyl-2-[methyl(methylsulfonyl)amino]pyrimidin-5-yl] (3R,5S)-3,5-dihydroxyhept-6- enoic acid or a pharmaceutically acceptable salt thereof and a second drug which is an inducer, inhibitor or substrate of P450 isoenzyme 4A4.
  17. 17. A pharmacy pack, as claimed in claim 16, wherein the second drug is used to lower cholesterol and is an inducer, inhibitor or substrate of P450 isoenzyme 3A4.
  18. 18. A pharmacy pack, as claimed in claim 17, wherein the second drug is selected from bezafibrate, clofibrate, fenofibrate, gemfibrozol and niacin.
  19. . 19. A pharmacy pack, as claimed in claim 16, wherein the second drug is used in treating cardiovascular conditions and is also an inhibitor, inducer or substrate of P450 isoenzyme 3A4,
  20. 20. A pharmacy pack, as claimed in claim 19, wherein the second drug is selected from, digitoxin, diltiazem, losartan, nifedipine, quinidine, verapimil and warfarin.
  21. 21. A pharmacy pack, as claimed in claim 16, wherein the second drug is a substrate of P450 isoenzyme 3A4 and is selected from acetominophen, aldrin, aflentanil, amiodorane, astemizole, benzphetamine, budenoside, carbamazepine, cyclophosphamide, cyclosporin, dapsone, digitoxin, ditiazem, diazepam, erthromycin, etoposide, flutamide, hydroxyarginine, ifosphamide, imipramine, lansoprazole, lidocaine, lovatidine, losartan, lovastatin, midrazolam, nifedipine, omeprazole, quinidine, rapamycin, retenoic acid, steroids, tacrolimus, teniposide, theophyline, toremifene, triazolam, troleandomycin, verapamil, warfarin, zatosetron and zonisamide.
  22. -44- PCT/GB00/00278 22, A pharmacy pack, as claimed in claim 16, wherein the second drug is an inhibitor of P450 isoenzyme 3A4 and is selected from clotrimazole, ethinylestradiol, gestodene, itraconazole, ketoconazole, miconazole, diltiazem, naringenin, erythromycin, cyclosporin and triacetyloleandomycin.
  23. 23. A pharmacy pack, as claimed in claim 16, wherein the second drug is an inhibitor of P450 isoenzyme 3A4 and is selected from carbamazepine, dexamethasone, phenobarbital, phenytoin, rifampin, sulfadimidine, sulfinipyrazone and triacetyloleandomycin.
  24. 24. Use of (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl) amino]pyrimidin-5-y1](3R,5S)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof in the medicament for use in combination therapy with a second drug which is an inducer, inhibitor or substrate of P450 isoenzyme 3A4.
  25. 25. Use of (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl) amino]pyrimidin-5-yl] (3R,5S)-3,5 dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof in the preparation of a medicament for use in cholesterol lowering therapy in combination therapy with a second drug which is an inducer, inhibitor or substrate of P450 isoenzyme 3A4.
  26. 26. Use, as claimed in claim 25, wherein the second drug is selected from bezafibrate, clofibrate, fenofibrate, gemfibrazol and niacin.
  27. 27. Use of (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino] pyrimidin-5-y1](3R,5S)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof in the preparation of a medicament for use in the treatment of cardiovascular condition in combination with a second drug which is an inducer, inhibitor or substrate of P450 isoenzyme
    3A4. AMENDED SHEET
    PCT/GB00/00278
  28. 28. Use, as claimed in claim 27, wherein the second drug is selected from digitoxin, diltuazam, losartan, nifedipine, quinidine, verapimil and warfarin.
  29. 29. Use of (E)-7-[4-(4-fluorophenyl)-6-1sopropyl-2- {methyl(methylsulfonyl)amino]pyrnimidin-5-y1] (3R,5S)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof in the preparation of a medicament for use in cholesterol lowering therapy in a patient receiving immunosuppresive therapy.
  30. 30. Use, as claimed in claim 29, wherein the immunosuppresive therapy comprises the administration of a drug selected from cyclosporin, tacrolimus and a corticosteroid.
  31. 31. Use of (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2- [methyl(methylsulfonyl)amino]pyrimidin-5-yl] (3R,55)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof in the preparation of a medicament for use in cholesterol lowering therapy in combination with a second drug which is selected from bezafibrate, clofibrate, fenofibrate, gemfibrazol and niacin in a patient receiving immunosuppresive therapy.
  32. 32. Use as claimed in claim 31 wherein the immunosuppressive therapy comprises the administration of a drug selected from cyclosporin, tacrolimus and a corticosteroid.
  33. 33. A substance or composition for use in a method of treatment for use in combination therapy with a second drug which is an inducer, inhibitor or substrate of P450 isoenzyme 3A4, said substance or composition comprising (E)-7-[4-(4- fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino]purimidin-5-y1}( 3R,5S5)-3,5- dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof, and said method comprising administering an effective amount of said substance or composition with said second drug. AMENDED SHEET
    -46- PCT/GB00/00278
  34. 34. A substance or composition for use in a method of treatment for use in cholesterol lowering therapy in combination therapy with a second drug which is an inducer, inhibitor or substrate of P450 isoenzyme 3A4, said substance or composition comprising (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methyl sulfonyl)amino]pyrimidin-5-y1](3R,5S)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof, and said method comprising administering an effective amount of said substance or composition with said second drug.
  35. 35. A substance or composition for use in a method of treatment as claimed in claim 34, wherein the second drug is selected from bezafibrate, clofibrate, fenofibrate, gemfibrazol and niacin.
  36. 36. A substance or composition for use in a method of treatment for use in the treatment of cardiovascular condition in combination with a second drug which is an inducer, inhibitor or substrate of P450 isoenzyme 3A4, said substance or composition comprising (E)-7-14-(4-fluorophenyl)-6-isopropyl-2-[methyl(methyl sulfonyl)amino]pyrimidin-5-yl]J(3R,5S)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof, and said method comprising administering an effective amount of said substance or composition with said second drug.
  37. 37. A substance or composition for use in a method of treatment as claimed in claim 36, wherein the second drug is selected from digitoxin, diltiazdm, losartan, nifedipine, quinidine, verapimil and warfarin.
  38. 38. A substance or composition for use in a method of treatment for use in cholesterol lowering therapy in a patient receiving immunosuppressive therapy, said substance or composition comprising (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2- [methyl(methylsolfonyl)amino]pyrimidin-5-y1](3R,5S)-3,5-dihydroxyhept-6-enoicacidor a pharmaceutically acceptable salt thereof, and said method comprising administering an effective amount of said substance or composition. AMENDED SHEET
    -47- PCT/GB00/00278
  39. 39. A substance or composition for use in a method of treatment as claimed in claim 38 wherein the immunosuppressive therapy comprises the administration of a drug selected from cyclosporin, tacrolimus and a corticosteroid.
  40. 40. A substance or composition for use in a method of treatment for use in cholesterol lowering therapy in combination with a second drug which is selected from bezafibrate, clofibrate, fenofibrate, gemfibrazol and niacin in a patient receiving immunosuppressive therapy, said substance or composition comprising (E)-7-[4-(4-fluorophenyl)-6-isopropyl- 2-[methyl(methylsulfonyl)amino]pyrimidin- 5-yl](3R,5S8)-3,5-dihydroxyhept-6-enoic acid or a pharmaceutically acceptable salt thereof, and said method comprising administering an effective amount of said substance or composition with said second drug.
  41. 41. A substance or composition for use in a method of treatment as claimed in claim 40 wherein the immunosuppressive therapy comprises the administration of a drug selected from cyclosporin, tacrolimus and a corticosteroid.
  42. 42. A combination as claimed in claim 1, substantially as herein described and illustrated.
  43. 43. A formulation as claimed in claim 12, substantially as herein described and illustrated.
  44. 44. A pack as claimed in claim 16, substantially as herein described and illustrated.
  45. 45. Use as claimed in any one of claims 24, 25, 27, 29 or 31, substantially as herein described and illustrated.
  46. 46. A substance or composition for use in a method of treatment as claimed in any one of claims 33 to 41, substantially as herein described and illustrated.
  47. 47. A new combination, a new formulation, a new pack, a new use of a compound as defined in claim 24 or of a pharmaceutically acceptable salt thereof, or a substance or composition for a new use in a method of treatment,substantially as herein described. AMENDED SHEET
ZA200105838A 1999-02-06 2001-07-16 Drug combinations comprising (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino] pyrimidin-5-YL] (3R,5S) -3,5-dihydroxyhept-6-enoic acid and an inhibitor inducer or substrate of P450 isoenzyme 3A4. ZA200105838B (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GBGB9902593.4A GB9902593D0 (en) 1999-02-06 1999-02-06 Drug combinations

Publications (1)

Publication Number Publication Date
ZA200105838B true ZA200105838B (en) 2002-10-16

Family

ID=10847179

Family Applications (1)

Application Number Title Priority Date Filing Date
ZA200105838A ZA200105838B (en) 1999-02-06 2001-07-16 Drug combinations comprising (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino] pyrimidin-5-YL] (3R,5S) -3,5-dihydroxyhept-6-enoic acid and an inhibitor inducer or substrate of P450 isoenzyme 3A4.

Country Status (2)

Country Link
GB (1) GB9902593D0 (en)
ZA (1) ZA200105838B (en)

Also Published As

Publication number Publication date
GB9902593D0 (en) 1999-03-24

Similar Documents

Publication Publication Date Title
US20090042911A1 (en) Drug combinations comprising (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino]pyrimidin-5-YL](3R,5S)-3, 5-dihydroxyhept-6-enoic acid and an inhibitor, inducer or substrate of P450 isoenzyme 3A4
Kosoglou et al. Ezetimibe: a review of its metabolism, pharmacokinetics and drug interactions
US20040072852A1 (en) Use of rosuvastatin (zd-4522) in the treatment of heterozygous familial hypercholesterolemia
JP2010155866A (en) Use of substituted cyanopyrrolidine and combination preparation containing them for treating hyperlipidemia and associated disease
US20050187204A1 (en) Medicinal composition for lowering blood lipid level
AU2001242985B2 (en) New combination of a betablocker and a cholesterol-lowering agent
JP2024507810A (en) Combination therapy of obisetrapib and ezetimibe for use in statin-intolerant patients suffering from hyperlipidemia or mixed dyslipidemia
ZA200105838B (en) Drug combinations comprising (E)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino] pyrimidin-5-YL] (3R,5S) -3,5-dihydroxyhept-6-enoic acid and an inhibitor inducer or substrate of P450 isoenzyme 3A4.
RU2233162C2 (en) Medicinal combination comprising (e)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl(methylsulfonyl)amino]pyrimidine-5-yl]-(3r,5s)-3,5-dihydroxyhept-6-enoic acid and inhibitor, inductor or substrate for p-450 3a4 isozyme
US20060252814A1 (en) Formulation comprising a betablocker and optionally a cholestrol-lowering agent
Shimpi et al. Bempedoic acid a novel drug used for the treatment of Hyperlipidaemia: A Review
HU226062B1 (en) Drug combinations comprising (e)-7-[4-(4-fluorophenyl)-6-isopropyl-2-[methyl-(methylsulfonyl)-amino]pyrimidin-5-yl](3r,5s)-3,5-dihydroxyhept-6-enoic acid and an inhibitor, inducer or substrate of p450 isoenzyme 3a4
US20090018199A1 (en) Methods of Administering 3, 3, 14, 14 Tetramethyl Hexadecane 1, 16 Dioic Acid
Davidson et al. Combination of pravastatin and probucol in the treatment of primary hypercholesterolemia
Jokubaitis et al. Clinical experience with fluvastatin, the first synthetic HMG-CoA reductase inhibitor
Cada et al. Rosuvastatin calcium