Title of Invention

A PHARMACEUTICAL PRODUCT FOR COUNTERACTING DAMAGE TO THE HEART AND A PROCESS FOR MANUFACTURING THE SAME

Abstract The invention relates to the utilization of benzazepine-N-acetic acid derivatives which contain an oxogroup in addition to the nitrogen atom in the $g(a) position and which are substituted in the third position by a 1-(carboxyalkyl)-cyclopentyl-carbonyl-amino radical and to their salts and biolabile esters for the prophylaxis and/or treatment of heart damages caused by cardiotoxic doses of medicaments or chemicals in large mammals and particularly human beings. The invention particularly relates to the prophylaxis and/or treatment of heart damages, especially myocardial damages, which may occur during cytostatic chemotherapy. The invention further relates to the utilization of the above-mentioned benzazepine-N-acetic acid derivatives for adjuvant treatment in therapies in which medicaments with oxidative-toxic side effects are used. The invention additionally relates to the production of medicaments suitable for said prophylaxis and/or treatment or adjuvant treatment.
Full Text FORM 2
THE PATENTS ACT 1970
[39 OF 1970]
8s
THE PATENTS RULES, 2003
COMPLETE SPECIFICATION
[See Section 10; rule 13]
"A PHARMACEUTICAL PRODUCT FOR COUNTERACTING DAMAGE TO THE HEART AND A PROCESS FOR MANUFACTUPJNG THE SAME"
SOLVAY PHARMACEUTICALS GMBH, a Dutch company, of Hans-Bockler-Allee 20, D-30173 Hannover, Germany,
The following specification particularly describes the invention and the
manner in which it is to be performed: GRANTED
28-10-2005




The present invention relates to a pharmaceutical product for counteracting damage to the heart and a process for manufacturing the same.
The present invention relates to the use of benzazepine-N-acetic acid derivatives which contain an oxo group in the a-position to the nitrogen atom and are substituted in the 3-positioh by a 1-(carboxyalkyl)-cyclopeniyI-carbonyl-amirio radical, and of their salts and biolabile esters, in particular for the prophylaxis and/or treatment of damage to the heart in larger mammals and in particular humans which is caused by oxidative-toxic, in particular cardiotoxic, doses of medicaments or chemicals, and for the preparation of medicaments suitable for this prophylaxis and/or treatment. Generally, the invention also relates to the use of the aforementioned benzazepine-N-acetic acid derivatives for adjuvant treatment in therapies in which medicaments having oxidative-toxic, and in particular cardiotoxic, side-effects are used. Preferably, the invention relates to the prophylaxis and treatment of damage to the heart, in particular to the myocardium, which may occur during cytostatic chemotherapy.
It is known that the cytostatic agents used in the chemotherapy of malignant tumours may have cardiotoxic properties as an unwanted side-effect Thus, some antibiotics are also used in cytostatic therapy which, owing to their general toxic properties, cannot be used for the treatment of bacterial infections. These include, for example, the anthracyclines isolated from streptomyces species, which are among the important more recent developments in the field of cytostatic agents. However, the clinical usability of the anthracyclines isjimited by their more or less greatly marked cardjotoxicity. The cardiotoxicity in this case is correlated to the total dose administered, and is frequently irreversible. Presumably, the heart damage and the cytostatic effects of these antibiotics are based at least in part on the membrane action thereof, by means of which the membrane fluidity and permeability is increased by the binding of the antibiotic to components of the cell membrane. Furthermore, oxidative damage may also be considered as an additional cause.
2

Typical antibiotics used in cytostatic therapy are the anthracyclines daunorubicin and the prodrug thereof, zorubicin, doxorubicin (adriamycin) and epirubicin, and the synthetic antibiotic mitoxantrone.
Benzazepine-N-acetic acid derivatives which contain an oxo group in the a-position to the nitrogen atom and are substituted in the 3-position by a 1-(carboxyalkyl)-cyclopentyl-carbonyl-amino radical, and their salts and biolabiie esters fall within the scope of protection of benzazepine-, benzoxazepine- and benzothiazepine-N-acetic acid derivatives described in German Patent Application DE 195 10 566, which contain an oxo group in the a-position to the nitrogen atom and are substituted in the 3-position by a 1-(carboxyalkyl)-cyclopentyl-carbonyl-amino radical, and which have NEP-inhibiting effects on the heart. The benzazepine-N-acetic acid compounds used here within the scope of the present invention may be prepared using the process described in DE 195 10 566.
It is an object of the invention to develop novel pharmaceutical preparations for the prophylaxis and/or treatment of damage to the heart which occurs in connection with the use of cardiotoxic doses of medicaments or chemicals.
According to the invention, compounds of the general formula I


R2OOC—CH-CR,—C—CO—NH
CH, COOR3

wherein
R1 stands for a phenyl lower-alkyl group which may optionally be substituted in the phenyl ring by lower alkyl, lower alkoxy or halogen, or for a naphthyl lower-alkyl group, R2 "is hydrogen or a group forming a biolabiie ester, and R3 is hydrogen or a group forming a biolabiie ester,
-3-


and physiologically compatible salts of the acids of Formula I are used for the preparation of pharmaceutical preparations for the prophylaxis and/or treatment of damage to the heart, in particular to the myocardium, induced by cardiotoxic doses of medicaments, in particular of cytostatic agents, preferably of cytostatic antibiotics, or chemicals, in larger mammals and humans.
Furthermore, the compounds of the above general formula I and of physiologically compatible salts of acids of Formula I are used for the preparation of pharmaceutical preparations for adjuvant treatment in larger mammals and humans in therapies in which medicaments having oxidative-cytotoxic, in particular oxidative-cardiotoxic, side-effects are used.
Where the substituents in the compounds of Formula I are or contain lower alkyl or alkoxy groups, these may be straight-chain or branched and contain, in particular, 1 to 4, preferably 1 to 2, carbon atoms and are preferably methyl or methoxy. Where the substituents contain halogen, fluorine, chlorine or bromine, preferably fluorine or chlorine, are particularly suitable.
In the radical R1 the lower alkylene chain may contain 1 to 4, preferably 1 to 2, carbon atoms. In particular, R1 is an optionally substituted phenethyl group which can optionally be substituted one or more times by halogen, lower alkoxy or lower alkyl, or is a naphthylethyl group.
The compounds of Formula I are optionally esterified dicarboxylic acid derivatives. Depending on the form of administration, biolabile monoesters, particularly compounds in which R2 is a group forming a biolabile ester and R3 is hydrogen, or dicarboxylic acids are preferred, the latter being particularly suitable for i.v. administration.
Suitable groups forming biolabile esters R2 and R3 are lower alkyl groups, phenyl or phenyl-lower-alkyl groups which are optionally substituted in the phenyl ring by lower alkyl or by a lower alkylene chain bonded to two adjacent carbon atoms, dioxolanylmethyl groups which are optionally substituted in the dioxolane ring by lower alkyl, or C2-C6-alkanoyloxymethyl groups optionally substituted on the oxymethyl group by lower alkyl. Where the group forming a biolabile ester R2 or R3 is lower alkyl, this may be a preferably unbranched alkyl group with 1 to 4, preferably 2, carbon atoms.
-k-

Where the group forming a biolabile ester is an optionally substituted phenyl lower-alkyl group, its alkylene chain may contain 1 to 3, preferably 1, carbon atoms. Where the phenyl ring is substituted by a lower alkylene chain, this may contain 3 to 4, particularly 3, carbon atoms. Phenyl, benzyl or indanyl are particularly suitable as phenyl-containing substituents R2 and/or R3. Where R2 and/or R3 are an optionally substituted alkanoyloxymethyl group, their alkanoyloxy group may contain 2 to 6, preferably 3 to 5, carbon atoms and is preferably branched and can be, for example, a pivaloyloxymethyl radical (= tert.-butylcarbonyloxymethyl radical).
Suitable physiologically compatible salts of dicarboxylic acids or monoesters of Formula I include their alkali metal, alkaline earth metal or ammonium salts, for example sodium or calcium salts or salts with physiologically compatible, pharmacologically neutral organic amines such as, for example, diethylamine or tert.-butylamine.
The compounds of Formula I contain two chiral carbon atoms, namely the carbon atom which is in the 3-position of the ring framework and bears the amide side-chain, and the carbon atom of the amide side-chain which bears the radical R\ The compounds can therefore exist in several optically active stereoisomeric forms or as a racemate. According to the present invention both the racemic mixtures and the isomerically pure compounds of Formula I may be used.
It has now surprisingly been found that the group of compounds of Formula I used according to the invention and their physiologically compatible salts of the acids, in addition to their previously-known NEP-inhibiting properties, also have the ability to counteract damage to the heart due to cardiotoxic substances (active substances, chemicals), in particular catabolic and anabolic processes (remodelling) such as those of myocardial hypertrophy and fibrous tissue growth, and thus exert a protective action against these cardiotoxic substances in the heart. The compounds of Formula I and their physiologically compatible salts of the acids thus have a preventive or damage-reducing and hence anti-cardiotoxic effect in relation to damage to the heart due to cardiotoxic substances, in humans and larger mammals. The compounds of Formula I, including their salts of acids and the biolabile esters thereof, are therefore suitable for the prophylaxis and/or treatment of damage to the heart, in particular to the myocardium, induced by cardiotoxic doses of medicaments or chemicals of widely-varying kinds. The substances causally responsible for damage to the heart, such as medicaments, may be of a diverse nature, e.g. the cytostatic agents used in the
-5-

J "
chemotherapy of malignant tumours, in particular cytostatic antibiotics. Furthermore, it was discovered in this connection that the group of compounds of Formula I used according to the invention very generally also exhibit antioxidative properties. These properties may result in advantageous cytoprotective and in particular cardioprotective effects, so that the compounds used according to the invention are suitable for adjuvant treatment in larger mammals and humans in therapies in which medicaments having oxidative-cytotoxic and in particular oxidative-cardiotoxic side-effects are used.
The anti-cardiotoxic action, i.e. the preventive or damage-reducing action directed against damage to the heart due to cardiotoxic substances, and the antioxidative effect of the compounds of Formula I used according to the invention was demonstrated in pharmacological tests in vivo on rabbits and rats each with adriamycin-induced cardiomyopathy. It was demonstrated by measuring the action of the substances on rabbits in relation to the inhibition or reduction of adriamycin-induced remodelling processes on the heart, and by measuring the antioxidant activity of the compounds on rats.
Description of the test methods
A) The tests were carried out on rabbits of both sexes having an initial body weight of 2.1 + 0.2 kg. The animals were divided into 3 groups:
1. untreated animals (= control animals, n = 20);
2. animals treated with adriamycin (+ placebo instead of test substance, n = 8);
3. animals treated with adriamycin and test substance (n = 8).
The test substance used was (3S,2R")-3-{1-[2"-(ethoxycarbonyl)-4"-phenyl-butyl]-cyclopentane-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-acetic acid, being representative of the substances of Formula I usable according to the invention.
Groups 2 and 3 were administered 1 mg/kg adriamycin i.v. twice a week for 4 weeks. Group 3 of rabbits having adriamycin-induced cardiomyopathy was administered a daily oral dose of the test substance (30 mg/kg body weight) for 4 weeks, starting on the first day of adriamycin treatment, with their food. Once the 4 weeks had ended, the hearts were isolated and weighed. Then they were fixed with formalin for later biochemical
6

1
investigations (hydroxyproline content of the heart tissue, measured with the HPLC amino acid analysis after Blankenship, D.T. et al., Aunal. Biochem. 178, 227 - 232, 1989 and Schuster, R., J. Chromatogr. 431, 271 - 284, 1989). Both the increase in heart weight in relation to body weight and in the hydroxyproline content in the heart tissue compared with normal values are indicators of remodelling processes taking place in the heart. The test results are compiled [in] Table I below.
Table I: Reduction of the cardiac remodelling processes caused by adriamycin by
the test substance in rabbits" hearts

Measured parameters Group 1:
Untreated animals, n = 20 (X ± SEM) Group 2:
Animals treated with adriamycin + placebo, n = 8 (X ± SEM) Group 3:
Animals treated with adriamycin + test substance, n = 8 (X ± SEM) % Effect of the test substance (group 3 v. 2)
Ratio of heart weights to body weight
(g) 2.01 ± 0.08 3.39 ±0.13"* 2.79 ± 0.08+ -17.7
Hydroxyproline content of the heart
(pg/ng) 6.66 ± 0.45 10.68 ±0.69*** 9.26 ±2.51 -13.3
SEM = Standard Error of the Mean *** p * p With this test method, the treatment with the test substance resulted in a statistically significant reduction in the heart/body weight ratio compared with control animals treated with adriamycin. Adriamycin treatment (group 2) increased the heart/body weight ratio (measured in g/kg) highly statistically significantly, by about 69%, compared with the untreated control group (group 1). If the test substance was administered in addition to adriamycin (group 3), the adriamycin-induced increase in the heart/body weight ratio was reduced statistically significantly by about 18% compared with the animals treated with placebos (group 2).
-7-

/
The left-ventricular myocardial hydroxyproline concentration, which is a measurement of cardiac fibrous tissue growth, was less in the animals treated with test substance (group 3) than in the control animals treated with adnamycin (group 2). Adriamycin treatment increased the myocardial hydroxyproline content (measured in ug/ng) of the heart highly statistically significantly, by about 60%, compared with the untreated control group (group 1). If the test substance was also administered in addition to adriamycin (group 3), the adriamycin-induced increase in the hydroxyproline content could be reduced by about 13% compared with animals treated with placebo (group 2). It can be concluded from the results that the remodelling process of the extracellular myocardial matrix is significantly reduced by administering the test substance.
B) The tests were carried out on male Wistar rats having an initial body weight of 229 to 277 g. The animals were divided into 4 groups:
1. untreated animals (= control animals, n = 19);
2. animals treated with adriamycin (+ placebo instead of test substance, n = 14);
3. animals treated with test substance (n = 11);
4. animals treated with adriamycin and test substance (n = 14).
The test substance used was (3S,2R")-3-{1-[2"-(ethoxycarbonyl)-4"-phenyl-butyl]-cyclopentane-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-acetic acid, being representative of the substances of Formula I usable according to the invention.
The animals of groups 2 and 4 were administered 15 mg/kg adriamycin intraperitoneally over a period of 2 weeks. The animals of group 4 were administered 30 mg/kg of the test substance daily for 2 weeks with their food, starting with the first day of adriamycin treatment. The animals of group 3 were likewise administered 30 mg/kg of the test substance daily for 2 weeks with their food (but without adriamycin).
Once the 2-weeks" treatment had ended, the animals were anaesthetised with pentobarbital (50 mg/kg i.p.) and venous blood samples were taken, from which plasma was obtained. The concentration of lipid-peroxides and the ferroxidase activity in the plasma was measured using the methods of Wong, S.H.Y. et al., Clin. Chem. 33, 214-220,1987 or Johnson, D.A. et al., Clin. Chem. .13, 142-150, 1967. Furthermore, the a-tocopherol concentration in the plasma was measured using the method of Catignani,

X
°>
G.L. and Bieri, J.G., Clin. Chem. 29, 708-712, 1983. The test results are compiled in Table II below.


Table II: Inhibition of the pro-oxidative action of adriamycin by the test substance in rats

Measured plasma parameters Group 1:
Untreated animals, n = 19 (X ± SEM) Group 2:
Animals treated with adriamycin + placebo, n =14 (X ± SEM) Group 3:
Animals treated with test
substance, n = 11 (X ± SEM) Group 4:
Animals treated
with adriamycin +
test substance, n
=14
(X ± SEM) % Effect of the test substance





Group 4 v. 2 Group 3 v. 1
a-Tocopherol (Vitamin E) (ug/dl) 337.0 ±21.0 338.2 ±28.0 657.8 ±21.0*** 407.9 ±33.0 +20.6 +95.2
Lipid peroxides
(measured as malonic
dialdehyde
thiobarbituric acid
adducts)
(umol/l) 1.939 ±0.085 4.476 ± 0.404*** 2.319 ±0.086* 3.030 ± 0.235+++ -32.3 +19.6
Ferroxidase activity (IU/1) 0.2698 ±0.0107 0.3289 ±0.317* 0.2545 ±0.0104 0.2534 ±0.0128+ -23.0 -5.7
SEM = Standard Error of the Mean * p *** p
The test substance exhibited directly antioxidative effects (e.g. increase in plasma a-tocopherol compared with the control animals and the animals treated with adriamycin), and inhibited the pro-oxidative action of adriamycin, which was demonstrated by a significant reduction in lipid oxidation and plasma ferroxidase activity compared with the control rats treated with adriamycin. Administering the test substance increased the a-tocopherol content (Vitamin E, measured in ug/dl) in the plasma of the test animals in group 3 statistically highly significantly, by about 95%, compared with the control animals (group 1). In the animals treated with adriamycin and test substance (group 4), a considerable increase in the a-tocopherol content in the plasma by about 21% was likewise noted, compared with the animals of group 2 (adriamycin + placebo). The concentration of lipid peroxides in the rat plasma (measured as malonic dialdehyde thiobarbituric acid adducts) increased statistically highly significantly, by about 131%, for the animals of group 2, which were treated with adriamycin, compared with the control group (group 1). If the test substance was administered in addition to adriamycin (group 4), the increase in the concentration of lipid peroxides in the plasma, induced by the adriamycin, could be reduced statistically highly significantly by about 32% compared with group 2 (adriamycin + placebo). The total activity of the ferroxidase (measured in IU/I) in the rat plasma increased statistically significantly in the group treated with adriamycin (group 2) by about 22% compared with the control group (group 1). If the test substance was administered in addition to adriamycin (group 4), the ferroxidase activity decreased statistically significantly by 23% compared with group 2 (adriamycin + placebo), and thus corresponded approximately to the ferroxidase activity which was determined for the control group (group 1).
It can be concluded from these test results that the pro-oxidative action of adriamycin plays a part in the cardiotoxicity caused by this substance, and that the test substance has a positive influence on this cardiotoxicity due to its anti-oxidative properties.
In view of their effect described above, the compounds of Formula I are suitable as medicaments for larger mammals and in particular humans for the prophylaxis and/or treatment of damage to the heart caused by damaging influences of cardiotoxic doses of medicaments and other chemical substances, such as in particular remodelling processes on the heart, such as myocardial hypertrophy or fibrous tissue growth. The compounds of the general formula I also have an advantageous antioxidative effect. This means that damaging oxidative influences of other medicaments, such as
-11-



cytostatic agents, can be reduced. The compounds of Formula I can thus be used as medicaments for adjuvant treatment in those therapies in which medicaments having oxidative-toxic and in particular cardiotoxic side-effects are administered. In this case, dicarboxylic acids of Formula I and their salts are expediently used in medicament forms for parenteral, particularly i.v., administration, and mono- or diesters of Formula I are expediently used in orally administered medicament forms. The doses to be used may differ between individuals and will naturally vary according to the nature of the condition to be treated, the substance used and the form of administration. For example, parenteral formulations will generally contain less active substance than oral preparations. Generally, however, medicament forms having an active substance content of 1 to 200 mg per individual dose are suitable for administration to larger mammals, in particular humans.
As therapeutic agents, the compounds of Formula I may be contained with conventional pharmaceutical adjuvants in pharmaceutical preparations such as tablets, capsules, suppositories or solutions. These pharmaceutical preparations may be prepared according to known methods, using conventional solid or liquid vehicles such as lactose, starch or talc, or liquid paraffins and/or using conventional pharmaceutical adjuvants, such as tablet disintegrating agents, solubilisers or preservatives.
The invention also relates to products which contain a medicament having cardiotoxic side-effects or a medicament having oxidative-cytotoxic or oxidative-cardiotoxic side-effects, in particular a cytostatic agent having cardiotoxic side-effects, and a compound of the above Formula I or a physiologically compatible salt of acids of Formula I as a combination preparation for simultaneous, separate or stage-wise application in therapy with the medicament having cardiotoxic side-effects. In particular, these products contain as cytostatic agent a cytostatic antibiotic and a compound of Formula I or a physiologically compatible salt of acids of Formula I as a combination preparation for simultaneous, separate or stage-wise application in cytostatic chemotherapy. Such products may for example contain a cytostatic antibiotic from the group consisting of the anthracyclines, mitoxantrone or a prodrug thereof as antibiotic. In this case, the anthracycline may be in particular daunorubicin, doxorubicin (adriamycin) or epirubicin or a prodrug thereof, preferably doxorubicin (adriamycin) or a prodrug thereof.
The following examples are intended to explain the invention in greater detail, but in no way limit its scope.
w?~

i
Examples 1 and 2 below describe pharmaceutical preparations according to the invention which contain an active substance of Formula I, and the preparation of such pharmaceutical preparations. The compounds of Formula I used according to the invention may be prepared for this purpose according to the methods described in the above German Patent Application DE 195 10 566.
Example 1:
Tablets containing (3S,2"R)-3-{1 -[2"-(ethoxycarbonyl)-4"-phenyl-butyl]-cyclopentan-1 -carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-aceticacid
Tablets were prepared with the following composition per tablet:
(3S,2"R)-3-{1-[2"-(ethoxycarbonyl)-4"-phenyl-butyl]-cyclopentan-1-carbonylamino}-
2,3,4,5-tetrahydro-2-oxo-1 H-1-benzazepine-1-acetic acid 20 mg
Corn starch 60 mg
Lactose 135 mg
Gelatine (as 10% solution) 6 mg
The active substance, the corn starch and the lactose were thickened with the 10% gelatine solution. The paste was comminuted and the resulting granules were placed on a suitable sheet and dried at 45°C. The dried granules were fed through a crushing machine and mixed with the following further adjuvants in a mixer:
Talc 5 mg
Magnesium stearate 5 mg
Corn starch 9 mg
and then compressed to form tablets of 240 mg.
Example 2:
Injection solution containing (3S,2"R)-3-[1-(2"-carboxy-4"-phenyl-butyl)-cyclopentan-1-carbonylamino]-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-aceticacid
An injection solution having the following composition per 5 ml was prepared:
-13-



(3S,2"R)-3-[1-(2"-carboxy-4,-phenyl-butyl)-cyclopentan-1-carbonylamino]-2,3,4,5-
tetrahydro-2-oxo-1H-1-benzazepine-1-aceticacid 10 mg
Na2HP04.7H20 43.24 mg
Na2HP04.2H20 7.72 mg
NaCl 30.0 mg
Purified water 4,948.0 mg
The solids were dissolved in water, the solution was sterilised and was poured into ampoules in portions of 5 ml each.
Example 3:
Preferred compounds of Formula I for use according to the invention for the preparation of mprtirampnts for the prophylaxis and/or treatment of damage to the heart which is caused by oxidative-toxic and in particular cardiotoxic doses of medicaments, in particular for adjuvant treatment in therapies with such medicaments, such as in cytostatic chemotherapy, are, for example, (including the salts of acids):
3-{1-[2"-(ethoxycarbonyl)-4"-phenyl-butyl]-cyclopentan-1-ca rbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-acetic acid tert. butyl ester.
3-{1-[2"-(ethoxycarbonyl)-4"-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1 H-1-benzazepine-1 -acetic acid.
(3S,2"R)-3-{1-[2"-ethoxycarbonyl)-4,-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-acetic acid tert. butyl ester.
(3S,2"R)-3-{1-[2,-(ethoxycarbonyl)-4"-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-acetic acid.
3-{1-[2"-(tert.-butoxycarbonyl)-4"-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-acetic acid tert. butyl ester.
3-[1-(2"-carboxy-4"-phenyl-butyl)-cyclopentan-1-carbonylamino]-2,3,4,5-tetrahydro-2-oxo-1 H-1-benzazepine-1-acetic acid.


3-{1-[2"-(tert. butoxycarbonyl)-4"-phenyl-butyI]-cyclopentan-1-carbonylamino}-2,3>4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-acetic acid benzyl ester.
3-[1-(2"-carboxy-4*-phenyl-butyl)-cyclopentan-1-carbonylamino]-2,3,4,5-tetrahydro-2-oxo-1 H-1-benzazepine-1-acetic acid benzyl ester.
3-{1 -[2"-(tert. butylcarbonyloxymethoxycarbonyl)-4"-phenyl-butyl]-cyclopentan-1 -carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1H-1-benzazepine-1-acetic acid benzyl ester.
3-{1-[2"-(pivaloyloxymethoxycarbonyl)-4"-phenyl-butyl]-cyclopentan-1-carbonylamino}-2,3,4,5-tetrahydro-2-oxo-1 H-1-benzazepine-1-acetic acid.


WE CLAIM



1. A pharmaceutical product for counteracting damage to the heart due to cardiotoxic substance said product comprising:
(I) doses of cytostatic agent of the kind as hereindescribed for causing cardiotoxic or oxidative-cytotoxic or oxidative-cardiotoxic side-effects; and
(II) 1 to 200 mg of a compound of the general formula I


r O
k=OOC-CH-CH,-C-CO-NH Y"">CH,—COOtf

wherein
R1 stands for a phenyl lower-alkyl group which may optionally be substituted in the phenyl ring by lower alkyl, lower alkoxy or halogen, or for a naphthyl lower-alkyl group,
R2 is hydrogen or a group forming a biolabile ester, and R3 is hydrogen or a group forming a biolabile ester,
(III) optionally a physiologically compatible salt of an acid of formula
I as a combination preparation for simultaneous, separate or step-wise application in therapy with the medicament having cardiotoxic or oxidative-cytotoxic or oxidative-cardiotoxic side-effects.
2. A product as claimed in claim 1, wherein the cytostatic agent is a cytostatic antibiotic.
3. A product as claimed in claim 2, wherein the cytostatic antibiotic is selected from the group consisting of the anthracyclines, mitoxantrone or a prodrug thereof.
4. A product as claimed in claim 3, wherein the anthracycline is
16

daunorubicin, doxorubicin (adriamycin) or epirubicin or a prodrug thereof.
A product as claimed in claim 4, wherein the anthracycline is doxorubicin (adriamycin) or a prodrug thereof
6. A process for preparing a pharmaceutical product for counteracting damage to the heart due to cardiotoxic substance said process comprising mixing:
(I) doses of cytostatic agent of the kind as hereindescribed for causing cardiotoxic or oxidative-cytotoxic or oxidative-cardiotoxic side-effects; and
(III) 1 to 200 mg of a compound of the general formula I

wherein
R1 stands for a phenyl lower-alkyl group which may optionally be substituted in the phenyl ring by lower alkyl, lower alkoxy or halogen, or for a naphthyl lower-alkyl group,
R2 is hydrogen or a group forming a biolabile ester, and
R3 is hydrogen or a group forming a biolabile ester,
(III) optionally a physiologically compatible salt of an acid of formula
I as a combination preparation for simultaneous, separate or step-wise application in therapy with the medicament having cardiotoxic or oxidative-cytotoxic or oxidative-cardiotoxic side-effects.
A process for the preparation of a combination product as claimed in claim 1 comprising a synergistic mixture of a medicament having
17

cardiotoxic or oxidative-cytotoxic or oxidative-cardiotoxic side-effects and a compound of general formula I



wherein
R1 stands for a phenyl lower-alkyl group which may optionally be substituted in the phenyl ring by lower alkyl, lower alkoxy or halogen, or for a naphthyl lower-alkyl group, R2 is hydrogen or a group forming a biolabile ester, and R3 is hydrogen or a group forming a biolabile ester, or a physiologically compatible salt of an acid of formula I, wherein an amount of the medicament having cardiotoxic or oxidative-cytotoxic or oxidative-cardiotoxic side-effects which induces such side-effects is mixed with 1 to 200mg of the compound of formula I or a physiologically compatible salt of an acid of formula I.

Dated this 5th day of February, 2002.


(RITUSHKApEGI) OF REMFRY 85 &AGAR
ATTORNEY FOR THE APPLICANTS

18

Documents:

in-pct-2002-00153-mum-cancelde pages (28-10-2005).pdf

in-pct-2002-00153-mum-claims(granted) (28-10-2005).pdf

in-pct-2002-00153-mum-claims(granted)-(28-10-2005).doc

in-pct-2002-00153-mum-correspondence (ipo)-(27-10-2005).pdf

in-pct-2002-00153-mum-correspondence(28-5-2005).pdf

in-pct-2002-00153-mum-form1(5-2-2002).pdf

in-pct-2002-00153-mum-form19(30-6-2004).pdf

in-pct-2002-00153-mum-form1a(24-10-2005).pdf

in-pct-2002-00153-mum-form1a(28-10-2005).pdf

in-pct-2002-00153-mum-form2(granted)-(28-10-2005).doc

in-pct-2002-00153-mum-form2-(granted) (28-10-2005).pdf

in-pct-2002-00153-mum-form3(24-10-2005).pdf

in-pct-2002-00153-mum-form3(28-10-2005).pdf

in-pct-2002-00153-mum-form3(5-02-2002).pdf

in-pct-2002-00153-mum-form5(28-10-2005).pdf

in-pct-2002-00153-mum-form5(5-02-2002).pdf

in-pct-2002-00153-mum-petition under rule137(24-10-2005).pdf

in-pct-2002-00153-mum-power of authrity (5-2-2002).pdf

in-pct-2002-00153-mum-power of authrity( 24-10-2005).pdf


Patent Number 210919
Indian Patent Application Number IN/PCT/2002/00153/MUM
PG Journal Number 43/2007
Publication Date 26-Oct-2007
Grant Date 15-Oct-2007
Date of Filing 05-Feb-2002
Name of Patentee SOLVAY PHARMACEUTICALS GMBH
Applicant Address HANS-BOCKLER-ALLEE 20, D-30173 HANNOVER, GERMANY
Inventors:
# Inventor's Name Inventor's Address
1 ZSUZSANNA ROZSA BESCSI KORUT 37-39, H-6722 SZEGED, HUNGARY
2 JULIUS GY PAPP NEMES TAKACS U. 10, H-6722 SZEGED,
3 DIRK THORMAHLEN TALWEG 3, D-31039 RHEDEN,
4 HARALD WALDECK OKERWEG 4 D, D-30916 ISERNHAGEN,
PCT International Classification Number A61K 31/55 ; A61K 45/06 ; A61K 31/70
PCT International Application Number PCT/EP00/06525
PCT International Filing date 2000-07-10
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 199 32 555.3 1999-07-13 Germany