WO2000027386A1 - Use of propionylcarnitine for the manufacture of a medicament for inhibiting smooth muscle cell proliferation - Google Patents

Use of propionylcarnitine for the manufacture of a medicament for inhibiting smooth muscle cell proliferation Download PDF

Info

Publication number
WO2000027386A1
WO2000027386A1 PCT/IT1998/000318 IT9800318W WO0027386A1 WO 2000027386 A1 WO2000027386 A1 WO 2000027386A1 IT 9800318 W IT9800318 W IT 9800318W WO 0027386 A1 WO0027386 A1 WO 0027386A1
Authority
WO
WIPO (PCT)
Prior art keywords
medicament
acid
carnitine
propionyl
useful
Prior art date
Application number
PCT/IT1998/000318
Other languages
French (fr)
Italian (it)
Inventor
Menotti Calvani
Original Assignee
Sigma-Tau Industrie Farmaceutiche Riunite S.P.A.
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 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. filed Critical Sigma-Tau Industrie Farmaceutiche Riunite S.P.A.
Priority to PCT/IT1998/000318 priority Critical patent/WO2000027386A1/en
Priority to AU12586/99A priority patent/AU1258699A/en
Priority to KR1020017005575A priority patent/KR100694004B1/en
Priority to EP99956317A priority patent/EP1128823A1/en
Priority to AU12946/00A priority patent/AU1294600A/en
Priority to CA002350071A priority patent/CA2350071A1/en
Priority to JP2000580616A priority patent/JP2002529408A/en
Priority to PCT/IT1999/000357 priority patent/WO2000027387A1/en
Publication of WO2000027386A1 publication Critical patent/WO2000027386A1/en
Priority to US09/849,369 priority patent/US20020091092A1/en
Priority to US10/356,547 priority patent/US20030220363A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/221Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin with compounds having an amino group, e.g. acetylcholine, acetylcarnitine

Abstract

The use of propionyl L-carnitine for the preparation of a medicament having inhibiting activity of the proliferation of smooth muscular cells of vascular wall is herein disclosed. Said medicament is useful for the treatment of vascular pathologies, such as atherosclerosis, hypertension, pulmonary hypertensions, restenosis after angioplasty.

Description

USE OF PROPIONYLCARNITINE FOR THE MANUFACTURE OF A MEDICAMENT FOR INHIBITING SMOOTH MUSCLE CELL PROLIFERATION
The present invention relates to the use of propionyl L-carnitine
and the pharmaceutically acceptable salts thereof for the preparation
of medicaments useful in the treatment of blood vessel pathologies.
Background of the invention.
A number of studies demonstrated that cell proliferation plays a
pivotal role in atherosclerosis, hypertension pathogenesis and
restenosis after angioplasty or coronary stenting (Ross, 1976;
Schwartz, 1990).
Many experimental studies, carried out on human
atherosclerotic plaques, demonstrated that cell proliferation is a
determining phenomenon both in the early phases and in the
progression of the plaque.
Further, proliferation of smooth muscle cells, which migrated to
intima from vascular tunica media, represents cell basis of coronaric
restenosis after rivascularization processes through angioplasty or
dilatation by means of a stent.
This drawback is the major limit to the application of
percutaneous rivascularization in patients affected by acute coronary
syndromes, since it is responsible for about 40% of post-surgical
failures (Holmes et al., 1984). Therefore, making available substances capable of controlling
the proliferation of smooth muscular cells of vessel wall is a goal of
primary importance in the prevention of restenosis after angioplasty,
as the proliferative phenomenon occurs in a determined timed
corresponding to the first weeks following the intervention.
Proliferation control in experimental atherosclerotic lesions has
been obtained with cytostatic drugs, such as etoposide (Llera-Moya et
al., 1992), with steroid hormones (Cavallero et al.; 1971; 1973; 1975;
1976), progestinic hormones (Spagnoli et al., 1990), dexamethasone
(Asai et al., 1993).
Smooth muscle cell proliferation is also inhibited by calcium
antagonist substances due both to a decrease of DNA synthesis, such
as in case of verapamil (Stein et al., 1987) and to the interference in
second messenger systems (cAMP), as demonstrated for nifedipine
(Cheung et al., 1987).
The treatment wάth ACE-inhibitors resulted in the control of the
growth of intima thickening (Powell, 1989).
Other in-vitro studies evidenced an antiproliferative effect on
cultured smooth muscular cells of rat aorta given by simvastatine, a
HMG-CoA reductase inhibitor, used as hypolipidemic agent (Corsini et
al., 1991). Further, some substances having triglyceridemia lowering
effect, such as fibrates, showed to be able to prevent the progression of
atherosclerotic lesions in the human (Olsson et., 1990). In a manner similar to what observed in neoplasia (Kerr, 1994),
phenomena of population decrease are observed to occur together with
cell proliferation in atherosclerotic population and/ or in intima
thickening (Gabbiani, 1995), thus suggesting that highly proliferative
cells go toward apoptosis and that modulation of the latter plays an
important role in atherosclerotic lesion genesis.
Using apoptosis inducing substances bears the risk to provoke
a generalised phenomenon, with possible side effects, which can be
even very severe, such as in the case of stem cells.
It has now been found that propionyl L-carnitine, thanks to its
unexpected proapoptotic effect, is endowed with a specific action of
control on smooth muscular cells of vessels.
Abstract of the invention.
It is an object of the present invention the use of propionyl L-
carnitine and the pharmacologically acceptable salts thereof for the
preparation of a medicament having inhibiting activity on smooth
muscular cells of blood vessel walls.
The most important and surprising advantage of the present
invention is that the administration of propionyl L-carnitine does not
imply toxic effects on bone marrow and in gut, which have a good
production of blood cellular elements and a very good turnover of
intestinal mucosa cells, respectively. This and other aspects of the present invention will be illustrated in detail, also by means of
examples.
Detailed description of the invention.
The present invention is based on the application of the
discovery that propionyl L-carnitine (hereinafter PLC) induces the
phenomenon of programmed death (apoptosis) in the cells. This effect
allows the treatment of blood vessel pathologies based on the
proliferation of smooth muscular cells of vessel walls, such as
pulmonary hypertension, atheroclerosis, hypertension, restenosis after
angioplasty.
Accordingly, a first aspect of the present invention relates to the
use of propionyl L-carnitine and the pharmacologically acceptable salts
thereof for the preparation of a medicament having inhibiting activity
on the proliferation of smooth muscular cells of vessel walls.
A further object of the present invention relates to the use of
propionyl L-carnitine and the pharmacologically acceptable salts
thereof for the preparation of a medicament useful for the treatment of
atherosclerosis.
Another object of the present invention is the use of propionyl L-
carnitine and the pharmacologically acceptable salts thereof for the
preparation of a medicament useful for the treatment of hypertension.
A fourth object of the present invention is the use of propionyl
L-carnitine and the pharmacologically acceptable salts thereof for the preparation of a medicament useful for the treatment of pulmonary
hypertension.
Still another aspect of the present invention is the use of
propionyl L-carnitine and the pharmacologically acceptable salts
thereof for the preparation of a medicament useful to prevent
restenosis after angioplasty.
The medicament according to the present invention can be obtained
admixing the active ingredient (propionyl L-carnitine or a
pharmacologically acceptable salt thereof) with excipients suitable for
formulation of compositions intended for enteral administration (in
particular the oral one) or parenteral administration (in particular
through intramuscular or intravenous route). All such excipients are
well known to persons skilled in the art.
As pharmaceutically acceptable salt of propionyl L-carnitine, it
is intended any salt thereof with an acid which does not give rise to
unwanted side effects. These acids are well known to the
pharmacologists and to the experts of pharmaceutical technology.
Non-limiting examples of said salts are chloride, bromide,
orotate, acid aspartate, acid citrate, acid phosphate, fumarate and acid
fumarate, lactate, maleate and acid maleate, acid oxalate, acid
sulphate, glucose phosphate, tartrate and acid tartrate.
Some examples of formulations in the form of unitary dosages
are given. (a) Formulation for tablets
A tablet contains:
Active ingredient
propionyl L-carnitine HC1 mg 500
Excipients
Microcrystalline cellulose mg 54.0
Polyvinylpyrrolydone mg 18.0
Crospovidone mg 30.0
Magnesium Stearate mg 15.0
Fumed silica mg 3.0
Hydroxypropylmethylcellulose mg 10.0
Poliethylene glycole 6000 mg 2.5
Titanium dioxide mg 1.8
Methacrylate copolymer mg 8.3
Talcum (triventilated) mg 2.4
(b) Formulation of intravenously injectable bottles
A bottle contains:
Active ingredient
Propionyl L-carnitine HC1 mg 300
Excipient
Mannitol mg 300
A solvent vial contains:
Sodium acetate 3Η2Q mg 390 Water for injectable F.U. q. s. to ml 5
The medicament prepared according to the present invention
will be administered in the form of pharmaceutical composition, which
can be prepared according to the general common knowledge of the
person skilled in the art.
Depending on the administration route appropriately chosen,
oral, parenteral or intravenous; the pharmaceutical composition will be
in the suitable form.
Examples of pharmaceutical compositions, wherein the
medicament according to the present invention is comprised, are the
solid or liquid oral forms, such as tablets, all types of capsules, pills,
solutions, suspensions, emulsions in the form of unitary or divided
doses, syrups, ready-to-use or extemporary drinkable unit doses.
Other examples are parenteral forms, injectable forms for
intramuscular, subcutaneous or intravenous administration.
Controlled or programmed release forms are also appropriate.
Dosages, posolgy and general therapeutic regimen will be
determined by the physician according to his knowledge, patient's
conditions and the pathology to be treated.
The association, whether co-administered in the same
medicament or separately (at the same time or subsequently) of PLC
with other active ingredients is also comprised in the present
invention. In a first preferred embodiment, the present invention relates to
restenosis after angioplasty.
According to this first preferred embodiment, the
pharmacological dose of PLC is such as not to exceed hematic
concentration of 100 mM.
The following example further illustrate the invention.
EXAMPLE
1
Wistar male rats, weighing between 270 and 290 mg, were used
for the experiments. The rats were anaesthetised with Nembutal i.p.
(35 mg/kg body weight) and the thoracic portion of aorta was
submitted to endothelium mechanical removal with Fogarty 2F balloon
probe (Baxter USA), according to the Baugartner and Studer
method(1966) with minor modification (Orlandi 1994). The animals
were randomized into 5 groups, each group is reported in Table 1.
Two groups were subjected to pharmacological treatment with
propionyl L-carnitine (PLC, 120 mg/Kg p.c. die), one group was treated
with an ACE-inhibitor (Enalapril, 1 mg/Kg p.c. die); the two remaining
groups were the control. Moreover, some non-balloonized animals were
used as blanks. Table 1
Figure imgf000011_0001
The animals were sacrificed 3 and 15 days after de-
endothelialization. Two hours before sacrifice, all the rats received i.v.
a Bromodeoxyuridine solution (BrDU) (45 mg/kg body weight) in order
to verify cell proliferation. One hour before sacrifice, some randomly
selected animals received 1 ml Blue Evans (1% in 0.9% NaCl solution)
in order to evaluate the degree of aorta disruption.
At sacrifice, the animals were anaesthetised with i.p. Nembutal
and perfused, after washing with isotonic saline containing 3%
Dextran 70, with buffered formalin for 20 minutes. Aortae were
isolated, slightly washed in saline and dissected longitudinally.
Carotid, heart and small intestine were also excised. All the organs
were post-fixed in the same fixative for 24 hours at room temperature.
Some aortic fragments were used for electronic microscopy. Aortae
were rolled up and included in paraffin. Serial sections having 5 μm thickness were stained with Hematoxylin-Eosine, Verhoeff-Van Gieson
and Movat's pentachromic and used for morphologic and
morphometric studies.
In some non-perfused animals, fragments of aortic tissue were
frozen in liquid nitrogen for the determination of tissular carnitines
and for subsequent studies of molecular biology.
Immunohystochemical staining
In order to put in evidence proliferating cells in damaged
arteries, serial sections in paraffin of aortae were deparaffined,
rehydrated, immersed in a 3% H2O2 solution for 20 minutes and
incubated with trypsine (0,05 M in Tris-HCl, pH 7.6) at 37°C. After
that, sections were treated with 2N HC1 at 37°C for 30 minutes,
washed with 0.1 M sodium tetraborate for 10 minutes, incubated with
normal horse serum (Vector) and subsequently with and-BrDU
monoclonal antibody (Ylem) for 1 hour. The preparates were then
reacted with biotilinated anti-mouse IgG (Vector) and the
Streptoavidine-ABC-POD complex (Ylem).
The reaction was evidenced by using diaminobenzidine (DAB) as
final chromogen. The count of positive nuclei for BrDU was made on
the total number of nuclei. Such count was blind-made by two
researchers separately. The difference between the two counts was
always lower than 0.5%. All data were analysed with the T Student's test. The differences
between the groups were considered to be significant for P<0.05.
Morphometric analysis
The entity of intima thickening after 15 days was evaluated on
Verhoeff-Van Gieson stained sections, using a grid overlapped on the
image, consisting of 400 points, 1 cm from each other.
The analysis was made on hystological preparates through a
Hamamatsu C3077 camera controlled by a Hamamatsu DVS 3000
image analyser and connected to a Polyvar-Reichert microscope.
Morphometric evaluation was made at XI 16 magnification. The
following parameters were evaluated a) relative volume of intima
referred to arterial wall; b) relative volume of tunica media referred to
arterial wall, by counting the overlapping points on the intima and
mean tunica.
3- 12 aorta sections were used at different level for each animal.
This number was a function of the structure sizes, according to Sach's
formula, showing the number of fields necessary to obtain a
statistically significant sample.
Ultrastructural studies
Small aorta samples were selected for electronic microscopy.
Aortae were post-fixed in osmium tetraoxide and embedded in EPON
812. Ultra thin sections were stained with uranyl acetate followed by lead citrate and examined using a Hitachi H-7100 FA transmission
electronic microscope.
Tissular and plasma carnitine assay
2-3 ml of blood samples were withdrawn from each animal
before mechanical de-endothelialization and at the time of sacrifice.
Plasma was separated by centrifugation (300 rpm) for 20 minutes and
frozen for plasmatic carnitine assay according to the Pace et al.
method.
Aorta wall samples were withdrawn from some non-perfused
animals, randomly selected from each group, frozen in liquid nitrogen
and stored at -80°C for the carnitine assay, according to the above
Pace et al. reference.
RESULTS
Ultrastructural studies
Small aorta samples were selected for electronic microscopy.
Aortae were post-fixed in osmium tetraoxide and embedded into EPOC
812. Ultra thin sections were stained with uranyl acetate followed by
lead citrate and examined through a Hitatchi H-7100 FA transmission
electron microscope.
Tissular and plasmatic carnitine assay
2-3 ml blood samples were withdrawn from each animal before
mechanical de-endothelialization and just before sacrifice. Plasma was separated by centrifugation (3000 rpm) for 20 minutes and frozen for
the plasmatic carnitine assay according to the Pace et al. method.
Samples of aortic wall were taken from some non-perfused
animals, randomly selected from each group, frozen in liquid nitrogen
and kept at -80° C for the assay of tissutal carnitines according to the
above-mentioned Pace et al. method.
RESULTS
Lesion morphology
3 days after the mechanical lesion, rat aortae did not show
significant hystological alterations, except the lack of endothelial cell
coating.
15 days after, remodelling of arteria could be observed for the presence
of an intima thickening (or neointima), consisting in round or
lengthened cells immersed in abundant extracellular matrix. Immune
hystochemical study put in evidence in particular the presence of
abundant smooth muscular cells (SMC) inside neointima.
Studies on proliferation
a) 3 days after de-endothelialization: the count of anti-BrDU
staining positive nuclei showed substantial differences between the
two groups examined. Quantitative analysis (Table 2) puts in
evidence that the number of BrDU-positive nuclei is significantly
lower in the tunica media in the PLC-treated animals, with respect
to controls (59.3% reduction against control, p<0.02). In both groups the distribution of BrDU-positive nuclei is more
concentrated in the lumen portion of mean tunica with respect to
the adventitia portion, with a 2: 1 ratio,
b) 15 days after de-endothelialization: Table 3 shows that in each
group the proliferation index of SMCs is significantly higher
(p<0.001) in the intima with respect to the tunica media. No
significant differences are observed in the number of BrDU-positive
nuclei, in the intima and tunica media, by comparing PLC,
Enalapril and control animals.
Morphometric analysis
As described in Table 3, after 15 days from endothelial lesion, the
intima relative volume is significantly lower, both in the PLC-treated
(31.11% reduction against control, p<0.02) and ACE-antagonist-
treated (26.14% reduction against control, p<0.01) animals against
control animals.
Table 2
In- vivo treatment with propionyl L-carnitine (PLC) on the proliferation
of smooth muscle cells of rat aorta after mechanical de-endothelia¬
lization: percentage of proliferating cell nuclei (anti-bromodeoxyuridine
positive) after 3 days (± s.e.m)
Figure imgf000017_0001
Table 3
In vivo treatment with propionyl L-carnitine (PLC) and with the ACE-
antagonist Enalapril on the proliferation of smooth muscular cells of
rat aorta after mechanical de-endothelialization: percentage of
proliferating cells (anti-bromodeoxyuridine positive) and percentage
ratio between intima volume and aorta wall volume after 15 days (±
s.e.m.) (preliminary results).
Figure imgf000018_0001
(a) intima vs tunica media: p<0.0001 ; (b) intima vs tunica media:
p<0.0001; (c) intima vs tunica media: p<0.001 ; (d) intima vol. /wall vs
controls: p<0.02; (e> intima vol/wall vs controls: p< 0.01 EFFECT OF PROPIONYL L-CARNITINE IN THE CONTROL OF
PROLIFERATION/APOPTOSIS.
In vitro experiments were carried out to evaluate the effect of
propionyl L-carnitine (PLC) on smooth muscular cells (SMC) isolated
from aortae of spontaneously hypertensive rats (SHR) and, as control,
on SMC isolated from normotensive rats (WKY).
These in vitro studies evidenced that PLC, when administered
during culture exponential growth phase, reduces cell growth,
evaluated as cell number/ml, as well as DNA synthesis, evaluated
through incorporation of trititated thymidine (Tab. 4 and 5).
Table 4: cell number/ ml at culture days 2, 3, 4 and 6
Figure imgf000019_0001
Table 5: tritiated thymidine incorporation at culture day 6
Figure imgf000019_0002
As a further characterisation of smooth muscular cells in the
presence of PLC, the percentage of apoptotic cells was measured both
in basal conditions and in oxidative stress conditions. Apoptosis
evaluation was carried out by counting the number of apoptotic cells
present on a total of 1000 cells, after specific DNA staining with
Hoechst 33258. The results of this experiment demonstrated that, in
SHR cultures, PLC determines a significant increase of apoptosis
percentage in basal conditions and that this increase is more evident
under stress conditions.
In WKY cultures, apoptosis percentage is negligible (tab. 6)
Table 6: apoptotic cell percentage in basal conditions and under
oxidative stress.
Figure imgf000020_0001
The behaviour observed in SHR smooth muscular cells might be
in some way related to the deregulated expression of c-myc, which
characterises spontaneously hypertensive rats (Negoro et al., 1988).
Moreover, it was observed that c-myc actively cooperates in inducing apoptosis subsequently to a proliferation stop (Bennet et al., 1993;
Bissonette et al., 1993), accordingly the data shown above suggest that
PLC anti-proliferative effect may be related to an interference with DNA
replication.
References:
Spagnoli L.G. Giorn. Arterioscl. 1983; 8: 117- 145
Asai K, Funaki C, Hayashi T. et al. Arterioscl. Thromb. 1993; 13: 892-
899
Baumgartner H.R., Studer A, Pathol. Microbiol. (Base) 29: 393-405,
1966
Bennet M.R., Evan G.I., Newby A.C., Circ. Res. 1994; 74:525-536
Bissonette R.P., Shi Y., Mahboubi A., Glynn J.M. and Green D.R, Curr.
Commun. Cell. Mol. Biol. 1993
Bonanno E., Ghibelli L., Coppola S., Spagnoli L.G., International
Conference on Cell Death in Human Pathology. Lecce, June 22-25, p.
54, 1995
Bouchaton-Piallat M.L., Gabbiani F., Desmouliere A. and Gabbiani G.
Am. J. Pathol. 146: 1059-1064, 1995
Cavallero C, De Lellis C, Di Tondo U. et al. In: Cavallero C, editor.
The arterial wall in atherogenesi. Padova: Piccin Medical Book, 1975:
25-42
Cavallero C, Di Tondo U. Mingazini L. et al., Ahero sclerosis 1973; 17:
49-62
Cavallero C, Di Tondo U., Mingazzini P. et al. Atherosclerosis 1976;
25: 145- 152
Cavallero C, Turolla E. and Ricevuti G., Atherosclerosis 1971; 13: 9- Cheung W.T., Shi M.N., Young J.D. et al., Biochem. Pharmacol. 1987;
36: 2183-2189
Clowes A.W., Schwartz S.M., Cir. Res. 56: 139-145, 1985
Corsini A., Raiteri M., Soma M. et al., Pharmacol. Res. 1991 ; 23: 173-
180
Curi R., Bond J.A., Calder P.C. and Newsholme E.A., Gen. Pharmac.
1993; 24, 591-597
Holmes D.R.J., Vliestra R.E., Smith H.C., Am. J. Cardiol. 1984; 77C-
81C.
Kerr J.F.R., Winterford CM., Harmon B.V., Cancer 73: 2013-2026,
1994
Llera-Moya M., Rothblat G.H., Glick J.M. et al., Arterioscler. Thromb.
1992; 12: 1363- 1370
Mauriello A., Sangiorgi G., Orlandi A., Schiaroli S., Pertumo S.,
Spagnoli L.G.,
Negoro N., Inariba H., Inoue T., Kanayama Y., Takeda T.
Olsson A.G., Ruhn G., Erikson U., J. Int. Med. 227: 381, 1 990
Orlandi A., Ehrlich H.P., Ropraz P. et al., Arterioscler. Thromb. 1994;
14: 982-989
Orlandi A., Ropraz P. and Gabbiani G., Exp Cell Res 1994; 214: 528-
536
Powell J.S., Clozel J-P., Muller R.K.M., Kuhn H., Hefti F., Hosang M.,
Baumgarner H., Science 245: 186-188, 1989 Ross R., Glomset J.A., N. Engl. J. Med. 295: 369-377, 1976
Schwartz S.M., Heimark R.L., Majesky M.V., Physiol. Rev. 70: 1177-
1209, 1990
Spagnoli L.G., Giorn. Arterioscl. 1983; 8: 117- 145
Spagnoli L.G., Orlandi A., Marino B., Mauriello A., De Angelis C,
Ramacci M.T., Atherosclerosis 1995; 114, 29-44
Spagnoli L.G., Orlandi A., Marino B. et al., Atherosclerosis 1995; 1 14:
29-44
Spagnoli L.G., Orlandi A., Mauriello A., et al., Pathol. Res. Pract. 1992;
4-5: 637642
Spagnoli L.G., Orlandi A., Mauriello A. et al. Atherosclerosis 1991; 89:
11-24.
Spagnoli L.G., Palmieri G., Mauriello A. et al., Atherosclerosis 1990;
82: 27-36
Spagnoli L.G., Sambuy Y., Palmieri G. et al., Artery 1985; 13: 187-198
Stein O., Halperin G., Stein Y., Arteriosclerosis 1987; 7: 585-592

Claims

1. Use of propionyl L-carnitine and pharmaceutically acceptable salts
thereof for the preparation of a medicament having inhibiting
activity of the proliferation of smooth muscular cells of vascular
wall.
2. Use according to claim 1, wherein said medicament is useful for the
treatment of atherosclerosis.
3. Use according to claim 1 , wherein said medicament is useful for the
treatment of hypertension.
4. Use according to claim 1, wherein said medicament is useful for the
treatment of pulmonary hypertension.
5. Use according to claim 1, wherein said medicament is useful for the
prevention of restenosis after angioplasty.
6. Use according to anyone of claims 1 to 5, wherein said salt of
propionyl L-carnitine is selected from the group consisting of
chloride, bromide, orotate, acid aspartate, acid citrate, acid
phosphate, fumarate and acid fumarate, lactate, maleate and acid
maleate, acid oxalate, acid sulphate, glucose phosphate, tartrate
and acid tartrate.
PCT/IT1998/000318 1998-11-11 1998-11-11 Use of propionylcarnitine for the manufacture of a medicament for inhibiting smooth muscle cell proliferation WO2000027386A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
PCT/IT1998/000318 WO2000027386A1 (en) 1998-11-11 1998-11-11 Use of propionylcarnitine for the manufacture of a medicament for inhibiting smooth muscle cell proliferation
AU12586/99A AU1258699A (en) 1998-11-11 1998-11-11 Use of propionylcarnitine for the manufacture of a medicament for inhibiting smooth muscle cell proliferation
KR1020017005575A KR100694004B1 (en) 1998-11-11 1999-11-09 Use of propiony L-carnitine for the preparation of a medicament capable of inducing apoptosis
EP99956317A EP1128823A1 (en) 1998-11-11 1999-11-09 Use of propionyl l-carnitine for the preparation of a medicament capable of inducing apoptosis
AU12946/00A AU1294600A (en) 1998-11-11 1999-11-09 Use of propionyl l-carnitine for the preparation of a medicament capable of inducing apoptosis
CA002350071A CA2350071A1 (en) 1998-11-11 1999-11-09 Use of propionyl l-carnitine for the preparation of a medicament capable of inducing apoptosis
JP2000580616A JP2002529408A (en) 1998-11-11 1999-11-09 Use of propionyl L-carnitine for the preparation of a medicament capable of inducing apoptosis
PCT/IT1999/000357 WO2000027387A1 (en) 1998-11-11 1999-11-09 Use of propionyl l-carnitine for the preparation of a medicament capable of inducing apoptosis
US09/849,369 US20020091092A1 (en) 1998-11-11 2001-05-07 Use of propionyl L-carnitine for the preparation of a medicament capable of inducing apoptosis
US10/356,547 US20030220363A1 (en) 1998-11-11 2003-02-03 Use of propionyl L-carnitine for the preparation of a medicament capable of inducing apoptosis

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IT1998/000318 WO2000027386A1 (en) 1998-11-11 1998-11-11 Use of propionylcarnitine for the manufacture of a medicament for inhibiting smooth muscle cell proliferation

Publications (1)

Publication Number Publication Date
WO2000027386A1 true WO2000027386A1 (en) 2000-05-18

Family

ID=11332914

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IT1998/000318 WO2000027386A1 (en) 1998-11-11 1998-11-11 Use of propionylcarnitine for the manufacture of a medicament for inhibiting smooth muscle cell proliferation

Country Status (2)

Country Link
AU (1) AU1258699A (en)
WO (1) WO2000027386A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006061341A1 (en) * 2004-12-10 2006-06-15 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Use of propionyl l-carnitine for the preparation of a high-dose medicament for the treatment of peripheral arterial disease
WO2008125714A1 (en) * 2007-04-16 2008-10-23 Universidad De Sevilla Use of l-carnitine for treating arterial hypertension
EP2147671A1 (en) * 2007-04-06 2010-01-27 Changzhou Hi-tech District Multiple Dimension Industry Technology Institute CO., LTD. The composition comprising l-carnitine or derivatives thereof and its use
WO2011004038A1 (en) * 2009-07-07 2011-01-13 Universidad De Sevilla Use of l-carnitine for preventing inflammatory cardiac damage associated with arterial hypertension

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4343816A (en) * 1979-02-12 1982-08-10 Claudio Cavazza Pharmaceutical composition comprising an acyl-carnitine, for treating peripheral vascular diseases
EP0681839A2 (en) * 1994-05-12 1995-11-15 Hirohiko Kuratsune A pharmaceutical preparation comprising an acylcarnitine
EP0793962A1 (en) * 1996-03-04 1997-09-10 Sigma-Tau Industrie Farmaceutiche Riunite S.p.A. Medicament containing a carnitine derivative for treating arteriosclerosis obliterans
US5786326A (en) * 1995-02-03 1998-07-28 Horwitz; Lawrence D. Method for the treatment of atherosclerosis and vascular injury by prevention of vascular smooth muscle cell proliferation

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4343816A (en) * 1979-02-12 1982-08-10 Claudio Cavazza Pharmaceutical composition comprising an acyl-carnitine, for treating peripheral vascular diseases
EP0681839A2 (en) * 1994-05-12 1995-11-15 Hirohiko Kuratsune A pharmaceutical preparation comprising an acylcarnitine
US5786326A (en) * 1995-02-03 1998-07-28 Horwitz; Lawrence D. Method for the treatment of atherosclerosis and vascular injury by prevention of vascular smooth muscle cell proliferation
EP0793962A1 (en) * 1996-03-04 1997-09-10 Sigma-Tau Industrie Farmaceutiche Riunite S.p.A. Medicament containing a carnitine derivative for treating arteriosclerosis obliterans

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
A.ORLANDI ET AL.: "Propionyl-L-carnitine exerts a specific control on serum triglyceride levels and plaque progression in hypercholesteremic aged rabbits", AGE, vol. 16, no. 3, 1993, pages 117, XP002101595 *
L.G.SPAGNOLI ET AL.: "Propionyl-L-carnitine prevents the progression of atherosclerotic lesions in aged hyperlipemic rabbits", ATHEROSCLEROSIS, vol. 114, no. 1, 1995, pages 29 - 44, XP002101594 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006061341A1 (en) * 2004-12-10 2006-06-15 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Use of propionyl l-carnitine for the preparation of a high-dose medicament for the treatment of peripheral arterial disease
EP2147671A1 (en) * 2007-04-06 2010-01-27 Changzhou Hi-tech District Multiple Dimension Industry Technology Institute CO., LTD. The composition comprising l-carnitine or derivatives thereof and its use
EP2147671A4 (en) * 2007-04-06 2010-04-14 Changzhou Hi Tech Distr Multip The composition comprising l-carnitine or derivatives thereof and its use
US7834056B2 (en) 2007-04-06 2010-11-16 Shuhua Gu Pharmaceutical composition for gout
WO2008125714A1 (en) * 2007-04-16 2008-10-23 Universidad De Sevilla Use of l-carnitine for treating arterial hypertension
ES2306610A1 (en) * 2007-04-16 2008-11-01 Universidad De Sevilla Use of l-carnitine for treating arterial hypertension
WO2011004038A1 (en) * 2009-07-07 2011-01-13 Universidad De Sevilla Use of l-carnitine for preventing inflammatory cardiac damage associated with arterial hypertension
ES2351753A1 (en) * 2009-07-07 2011-02-10 Universidad De Sevilla Use of l-carnitine for preventing inflammatory cardiac damage associated with arterial hypertension

Also Published As

Publication number Publication date
AU1258699A (en) 2000-05-29

Similar Documents

Publication Publication Date Title
US5599844A (en) Prevention and treatment of pathologies associated with abnormally proliferative smooth muscle cells
US20060009397A1 (en) New use
CN103830719A (en) Methods for performing coronary artery bypass graft procedure
US6191144B1 (en) Method of using angiotensin converting enzyme inhibitor to stimulate angiogenesis
JP2003519183A (en) Use of low-dose bisphosphonates to inhibit cardiac and arterial calcification
WO2000027386A1 (en) Use of propionylcarnitine for the manufacture of a medicament for inhibiting smooth muscle cell proliferation
WO1997015598A1 (en) Novel peptide
HU206186B (en) Process for producing pharmaceutical compositions comprising amino acid derivatives having ace-inhibiting effect
EP0766963B1 (en) Arteriosclerosis depressant
US10172914B2 (en) Combination
US20020091092A1 (en) Use of propionyl L-carnitine for the preparation of a medicament capable of inducing apoptosis
US6019104A (en) Method for the treatment or prevention of restenosis associated with coronary intervention
Churchill et al. Enalapril in the prevention of restenosis following intracoronary intervention in a swine model
EP1128823A1 (en) Use of propionyl l-carnitine for the preparation of a medicament capable of inducing apoptosis
JPH07145129A (en) New application of beta-naphthoquinone derivative and its salt
JPH09151137A (en) Medicine for inhibiting multiplication of smooth muscle cell
JPH03500660A (en) Vasoocclusion inhibitor
WO2001041796A1 (en) Method of combating undesirable hyperplastic cell proliferation
JPH06157311A (en) Medicine for improvement of vessel anomaly
CN101219204A (en) Combinations comprising a low molecular weight thrombin inhibitor and a prodrug of a low molecular weight thrombin inhibitor
JPWO2003004036A1 (en) Pharmaceutical composition for treatment of secondary hyperparathyroidism and therapeutic agent for cardiovascular complications associated with treatment of secondary hyperparathyroidism
KR20050092519A (en) COMBINATIONS OF ATORVASTATIN AND α1 ADRENERGIC RECEPTOR ANTAGONISTS
JP2005320247A (en) Prophylactic or therapeutic agent for disseminated intravascular coagulation syndrome

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

NENP Non-entry into the national phase

Ref country code: CA

122 Ep: pct application non-entry in european phase