Find information on thousands of medical conditions and prescription drugs.

Carvedilol

Carvedilol (Coreg®) is a non-selective beta blocker indicated in the treatment of mild to moderate congestive heart failure (CHF). In addition to blocking both β1 and β2 type adrenoreceptors, carvedilol also displays α1-adrenergic antagonism as well, which confers the added benefit of reducing blood pressure through vasodilation. more...

Home
Diseases
Medicines
A
B
C
Cabergoline
Caduet
Cafergot
Caffeine
Calan
Calciparine
Calcitonin
Calcitriol
Calcium folinate
Campath
Camptosar
Camptosar
Cancidas
Candesartan
Cannabinol
Capecitabine
Capoten
Captohexal
Captopril
Carbachol
Carbadox
Carbamazepine
Carbatrol
Carbenicillin
Carbidopa
Carbimazole
Carboplatin
Cardinorm
Cardiolite
Cardizem
Cardura
Carfentanil
Carisoprodol
Carnitine
Carvedilol
Casodex
Cataflam
Catapres
Cathine
Cathinone
Caverject
Ceclor
Cefacetrile
Cefaclor
Cefaclor
Cefadroxil
Cefazolin
Cefepime
Cefixime
Cefotan
Cefotaxime
Cefotetan
Cefpodoxime
Cefprozil
Ceftazidime
Ceftriaxone
Ceftriaxone
Cefuroxime
Cefuroxime
Cefzil
Celebrex
Celexa
Cellcept
Cephalexin
Cerebyx
Cerivastatin
Cerumenex
Cetirizine
Cetrimide
Chenodeoxycholic acid
Chloralose
Chlorambucil
Chloramphenicol
Chlordiazepoxide
Chlorhexidine
Chloropyramine
Chloroquine
Chloroxylenol
Chlorphenamine
Chlorpromazine
Chlorpropamide
Chlorprothixene
Chlortalidone
Chlortetracycline
Cholac
Cholybar
Choriogonadotropin alfa
Chorionic gonadotropin
Chymotrypsin
Cialis
Ciclopirox
Cicloral
Ciclosporin
Cidofovir
Ciglitazone
Cilastatin
Cilostazol
Cimehexal
Cimetidine
Cinchophen
Cinnarizine
Cipro
Ciprofloxacin
Cisapride
Cisplatin
Citalopram
Citicoline
Cladribine
Clamoxyquine
Clarinex
Clarithromycin
Claritin
Clavulanic acid
Clemastine
Clenbuterol
Climara
Clindamycin
Clioquinol
Clobazam
Clobetasol
Clofazimine
Clomhexal
Clomid
Clomifene
Clomipramine
Clonazepam
Clonidine
Clopidogrel
Clotrimazole
Cloxacillin
Clozapine
Clozaril
Cocarboxylase
Cogentin
Colistin
Colyte
Combivent
Commit
Compazine
Concerta
Copaxone
Cordarone
Coreg
Corgard
Corticotropin
Cortisone
Cotinine
Cotrim
Coumadin
Cozaar
Crestor
Crospovidone
Cuprimine
Cyanocobalamin
Cyclessa
Cyclizine
Cyclobenzaprine
Cyclopentolate
Cyclophosphamide
Cyclopropane
Cylert
Cyproterone
Cystagon
Cysteine
Cytarabine
Cytotec
Cytovene
Isotretinoin
D
E
F
G
H
I
J
K
L
M
N
O
P
Q
R
S
T
U
V
W
X
Y
Z

More importantly, carvedilol also has a minimal potential for "inverse agonism", or the deactivation of an activated receptor. This is important to CHF sufferers since inverse agonism causes negative chronotropic and inotropic effects. Essentially, carvedilol does not decrease the rate or strength of the hearts contractions as much as other beta blocking medications. CHF often significantly reduces how well the heart pumps, so any medication that further weakens the rate or strength of contractions is undesireable, therefore making carvedilol a better treatment than a beta blocker with stronger inverse agonism (such as propranolol).

On January 10, 2006, GlaxoSmithKline announced to pharmicists and physicans that there will be a limited availability of Coreg. This is due to documentation procedures with the manufacturer. It is not known when will Coreg will become broadly available. Patients who are taking Coreg should consult their healthcare professional about what actions they should take due to the shortage.

Read more at Wikipedia.org


[List your site here Free!]


Antioxidant properties of carvedilol: Inhibition of lipid peroxidation, protein oxidation and superoxide generation
From Neurological Research, 10/1/03 by Suzuki, Yoko

Oxidative stress has been suggested to be an etiological factor in cerebro- and cardiovascular disorders. We examined antioxidant activities of carvedilol, a [beta]- and [alpha]-adrenoreceptor blocker. Carvedilol suppressed lipid auto-oxidation and protein carbonyl formation in brain homogenate in a dose-dependent manner. Carvedilol also suppressed superoxide generation of human neutrophils. These properties of carvedilol should be suitable for treating hypertension resulting in cerebro- and cardiovascular diseases. [Neurol Res 2003; 25: 749-753]

Keywords: Carvedilol; antioxidant; lipid peroxidation; protein carbonyls; superoxide

INTRODUCTION

Oxidative stress has been proposed as an important pathologic factor of ischemia-reperfusion and age-associated vascular pathologies such as arteriosclerosis1,2. In addition, oxidative stress is considered to play a cytotoxic role in age-related neurodegenerative disorders including Parkinson's and Alzheimer's diseases2-4. Neuronal cells are extremely sensitive to injury by reactive oxygen species that mediate cell death in various pathologic conditions. Therefore, agents with antioxidant activity as well as antihypertensive activity should be more beneficial in treating hypertension. Hypertension is the most critical risk factor of cerebrovascular diseases and is often associated with them.

Carvedilol, (+ or -)-1-(carbazol-4-yloxy)-3-[2-methoxyphenoxy] ethylamino-2-propanol, has been used clinically to treat hypertension for several years. The clinical effect is mainly derived from [beta]- and [alpha]-adrenoreceptor blockade. In addition, carvedilol has been shown to act as an antioxidant5-2. Several studies reported the antioxidative effects of carvedilol, using different systems for radical generation including auto-oxidation of dihydroxyfumaric acid in the presence or absence of iron ions7,8, ferrous iron with ascorbic acid9, stimulated macrophages5,6, and enzymatic systems9. However, the mechanism of this antioxidative effect is not completely clear.

To investigate whether carvedilol has neuroprotective ability, we examined its antioxidant properties by three independent approaches: 1. auto-oxidation of brain homogenate measured as thiobarbituric acid-reactive substances, 2. protein oxidation of brain homogenate monitored by protein carbonyl formation, and 3. superoxide-generation from neutrophils.

MATERIALS AND METHODS

Effects on lipid peroxidation

A whole swine brain was rapidly removed after slaughter. The cerebral cortex was collected and stored at -80[degrees]C until use.

The cerebral cortex was homogenized in modified Hank's balanced salt solution (mHBSS, Nissui, Japan) without phenol red and sodium bicarbonate, adjusted to pH 7.4 by addition of 280 mOsm disodium hydrogen phosphate. To induce auto-oxidation, the 5% homogenate with or without carvedilol was incubated at 37[degrees]C. Lipid peroxides produced by auto-oxidation were monitored by levels of thiobarbituric acid-reactive substances (TBARS).

After the incubation period, 500 [mu]l of the reaction mixture was added to 2 ml TBA reagent consisting of 15% trichloroacetic acid (Wako Pure Chemical, Osaka, Japan), 0.375% TBA, 0.25 M HCl dissolved in distilled water, 30 [mu]l butylated hydroxytoluene (BHT; 50 mM ethanol solution) and 470[mu]l distilled water, was heated in a 100[degrees]C water bath for 15 min. TBA and BHT were purchased from Tokyo Kasei (Japan) and Wako Pure Chemical (Osaka, Japan), respectively. After cooling in ice-cold water, the mixture was centrifuged at 1,250xg for 10 min. The absorbance of the supernatant at 535 nm was read against a blank which contained 2 ml of TBA reagent, 30 [mu]l BHT and 970 [mu]l of distilled water, using a spectrophotometer (U-2000, Hitachi, Tokyo, Japan). The concentration of TBARS was calculated using a molar absorption coefficient of 156,000 M cm^sup -1^.

Under the same conditions, anti-oxidant properties of carvedilol were compared with those of [alpha]-tocopherol (a kind gift of Eisai, Tokyo, Japan).

Effects on protein oxidation

Sample preparation

After the whole brain was rapidly removed from a male Wistar rat, the cerebral cortex was collected and stored at -80[degrees]C until use. The cerebral cortex (50 mg) was homogenized in 0.55 ml of buffer in a glass homogenizer. The buffer consisted of 0.5 ml of homogenizing buffer (0.1% digitonin in 100 mM potassium dihydrogen orthophosphate buffer; PDO buffer, pH 7.4), 50 [mu]l of protease inhibitor cocktail (5 [mu]g ml^sup -1^ aprotinin, 5 [mu]g ml^sup -1^ leupeptin, 7 [mu]g ml^sup -1^ pepstatin A and 10 mM EDTA), and 1 [mu]l of phenylmethylsulphonyl fluoride (40 mg ml^sup -1^ dissolved in ethanol). Digitonin, aprotinin, leupeptin, pepstatin A, EDTA, and phenylmethylsulphonyl fluoride were purchased from Sigma Chemicals, St. Louis, MO, USA. The homogenized tissue was left at room temperature for 15 min and then centrifuged at 2,755xg for 10 min. The supernatant was removed and incubated at room temperature for 15 min with 1% streptomycin sulfate solution. Centrifugation at 2,755xg for 10 min removed any nucleic acids that might interfere with the carbonyl assay. The supernatant was used as the sample.

Protein oxidation induced by exposure to hydrogen peroxide

Bovine serum albumin (BSA; Sigma Chemicals) dissolved in PDO buffer (0.5 mg ml^sup -1^) and the homogenate of rat cerebral cortex were exposed to 0.2 mM hydrogen peroxide (H^sub 2^O^sub 2^; Kanto Chemical, Tokyo, Japan) in 100 mM PDO buffer, and then incubated for 20 min at 37[degrees]C. Carvedilol at various concentrations was added to the basal system before H^sub 2^O^sub 2^ exposure. To remove H^sub 2^O^sub 2^, manganese oxide was added. After centrifugation twice at 10,000xg for 5 min, the supernatant was used as the protein sample.

Derivatization of proteins with 2,4-dinitrophenylhydrazine (2,4-DNPH)

Using the OxyBlot(TM) Protein Oxidation Detection Kit (Intergen, Purchase, NY, USA) and according to its protocol, protein carbonyls were derivatized with 2,4-DNPH. Briefly, after denaturation of 5 [mu]l of protein samples by adding 5 [mu]l of 12% SDS, the samples were derivatized by adding 10 [mu]l of 1xDNPH solution and then incubated for 15 min at room temperature. Finally, 7.5 [mu]l of neutralization solution and 1.6 [mu]l of 2-mercaptoethanol were added. Derivatized samples were assayed within 24 h.

Detection of derivatized proteins by Western blot

Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) was carried out by standard procedures using the Mini Protean II system (Bio-Rad, Hercules, CA, USA). Samples treated with DNPH (derivatized BSA and brain homogenate) and molecular weight protein standards (OxyBlot(TM) Protein Oxidation Detection Kit; Intergen) were loaded on a 10% acrylamide resolving gel with a 4% acrylamide stacking gel. After electrophoresis, proteins were transferred to a PVDF membrane (Amersham Pharmacia, Buckingham, UK) using the Trans-Blot SD system (Bio-Rad) at 15 V for 20 min. The membranes were blocked for 1 h with 5% nonfat milk powder in PBS-T consisting of phosphate buffer saline supplemented with 0.1% Tween 20 (Wako Pure Chemical) and were then incubated overnight with rabbit anti-DNP antibody diluted with 1% nonfat milk powder in PBS-T (1 : 150). After washing with PBS-T (3x30 min), blots were incubated with a 1 : 300 dilution in PBS-T of secondary goat anti-rabbit antibody conjugated with horseradish peroxidase for 1 h. After washing (3x30 min with PBS-T), the blots were visualized using a chemiluminescence reagent, ECL-PLUS (Amersham). Finally, the membranes were exposed to Hyperfilm-ECL (Amersham).

Semi-quantitative analysis for protein carbonyls by densitometry with dot blot

Aliquots of 3 [mu]l of sample treated with DNPH were applied to a PVDF membrane. After the membranes were dried, they were blocked with 5% nonfat milk powder in PBS-T. The following procedures were the same as the Western blot assay mentioned above. Optical densities of the immunoblots or total protein carbonyl content were compared. The density of each band was measured using image processing software, NIH image (NIH, Bethesda, MD, USA).

Effects on superoxide production

Xantine oxidase (from buttermilk, grade III) and phorbolmyristate acetate (PMA) were purchased from Sigma Chemicals. PMA was dissolved in a small amount of dimethyl sulfoxide (DMSO; Sigma Chemicals), diluted with 50% DMSO and 50% mHBSS to the specified concentration, and then stored at -80[degrees]C. Hypoxantine was obtained from Wako. 2-Methyl-6- (p-methoxyphenyl)-3,7dihydroimidazo [1,2-a] pyrazin-3-one (MCLA) was purchased from Tokyo Kasei and prepared by the method described previously13.

Blood was drawn into a heparinized syringe (20 units ml^sup -1^) from healthy volunteers. Leukocytes were isolated by sedimentation in the presence of dextran followed by brief hypotonic lysis of contaminating erythrocytes13. The leukocytes, which contained 60%-80% neutrophils, were suspended in mHBSS and kept at 4[degrees]C for no longer than 3 h prior to use.

Superoxide production was determined by the MCLA dependent chemiluminescence method using a BLR 301 Luminescence Reader (Aloka, Tokyo, Japan)14. All experiments were carried out in the incubation chamber of the Luminescence Reader at 37[degrees]C in a total volume of 1 ml. The reaction mixture for the basal system consisted of neutrophils (5.0x10^sup 4^ cells ml^sup -1^) and 1.0 [mu]M MCLA in mHBSS with pre-incubation for 5 min at 37[degrees]C. The reaction was started by the addition of 0.1 [mu]g ml^sup -1^ of PMA. Superoxide production was expressed as photon counts per min. The maximum photon count was defined as the peak count14. The incubation period was 5 min. Carvedilol at various concentrations was added to the basal system before pre-incubation.

The hypoxanthine-xanthine oxidase system was used to test the scavenging effect of carvedilol on superoxides and the possible interaction with MCLA as described previously15. A system containing 1.0 [mu]M MCLA, 0.18 mg ml^sup -1^ of BSA, 45 [mu]M hypoxanthine and 0.1% N, N-dimethylformamide (Sigma Chemicals) in mHBSS was incubated with or without 100 [mu]M carvedilol at 37[degrees]C. Chemiluminescence produced in this system by addition of xanthine oxidase (28 units ml^sup -1^) was monitored.

RESULTS

Effects on lipid peroxidation

TBARS formation was increased with increasing incubation time (Figure 1). The addition of 25 [mu]M carvedilol significantly (about 50% inhibition) suppressed TBARS formation. Comparison between the two groups using two-factor analysis of variance (ANOVA) (one factor for the groups with or without carvedilol and another for the incubation time) revealed significant differences in the groups (p

Effects on protein oxidation

Carvedilol strongly inhibited protein carbonyl formation both in BSA and rat brain homogenate (Figure 3). Carvedilol (100-200 [mu]M) significantly suppressed protein carbonyl formation. The inhibitory effect was found to be dose-dependent (Figure 4). In the Western blot assay, carvedilol had different antioxidant effects on each protein band (Figure 3).

Effects on Superoxide production

Carvedilol suppressed the chemiluminescence of neutrophils stimulated by PMA and reduced the peak count in a dose-dependent manner (Figure 5). Complete inhibition was achieved by addition of 150 [mu]M carvedilol. The concentration required for 50% inhibition of the peak count was about 60 [mu]M.

In all experiments using the MCLA-dependent chemiluminescence method, the complete scavenging effect of 0.5 [mu]M SOD revealed that the chemiluminescence was derived from Superoxide15. In the hypoxanthinexanthine oxidase system, chemiluminescence produced in the presence of 100 [mu]M carvedilol was essentially the same as that shown in the system without carvedilol (data not shown). Therefore, carvedilol itself showed no detectable superoxide-scavenging activity and no interference with MCLA, but had an inhibitory effect on the superoxide-generating system of neutrophils.

DISCUSSION

We demonstrated that carvedilol has antioxidative ability by three independent approaches; lipid peroxidation, protein oxidation, and Superoxide production.

In the present study, carvedilol markedly inhibited lipid auto-oxidation of rat brain homogenates. This result agrees with those of previous studies5. Its inhibition of brain auto-oxidation is significantly more pronounced than that of [alpha]-tocopherol, a noble antioxidant breaking free-radical chain reactions in biological membranes. Carvedilol is a lipophilic agent which can be easily incorporated into membranes and is able to cross the blood-brain barrier. Although the mechanism of carvedilol inhibition of auto-oxidation of brain homogenate has not been elucidated, its lipid solubility is advantageous for antioxidant activities that inhibit membrane lipid peroxidation because initiation and propagation of radical chain reactions prefer lipophilic conditions. Carvedilol may act as an antioxidant breaking radical chain reactions by scavenging peroxyl and alkoxyl radicals, which are abundantly generated by iron-dependent decomposition of preformed lipid peroxides16. Our results suggest that this effect of carvedilol may protect biologic membranes from oxidative stress.

We also showed marked antioxidant activity of carvedilol in protein oxidation. Many studies suggest that oxidative stress is responsible for age associated pathologies such as arteriosclerosis and neurodegenerative disorders2-4. Oxidation of proteins modifies the side chains of methionine, histidine and tyrosine, forming cystein disulfide bonds17. Metal catalyzed oxidation of proteins introduces carbonyl groups (aldehydes and ketones) in lysine, arginine, proline or threonine residues in a site-specific manner17. Many investigators have demonstrated that hydroxyl radical causes oxidative modification of amino acid residues of proteins and that cross-linking and fragmentation of the proteins result in loss of function and increased susceptibility to proteases18-21. However, there have been no reports showing inhibitory effects of carvedilol on protein oxidation in the brain. We demonstrated this property for the first time. The inhibitory effect of carvedilol on protein oxidation is not clear, but it may inhibit production of the hydroxyl radical. Our findings suggest that carvedilol may protect against cellular and extracellular protein damage from oxidative stress and permit maintenance of physiological function.

We demonstrated that carvedilol did not scavenge Superoxide, but suppressed the superoxide-generating system. Since Superoxide is a potential precursor of the hydroxyl radical, which seems to be a more hazardous free radical, carvedilol may indirectly prevent hydroxyl radical formation by inhibition of Superoxide production from neutrophils. Recently, neutrophils have been suggested to play a role in ischemia/reperfusion injury. The removal of circulating neutrophils is reported to reduce ischemic brain edema22. Superoxide produced by neutrophils is one of the critical chemical mediators that alter vascular permeability and accelerate the development of cerebral edema22,23. Our findings suggest that carvedilol may have a protective effect on brain tissue during the acute phase of ischemia/reperfusion injury by suppression of superoxide production from neutrophils, thus reducing the size of infarction.

CONCLUSION

We demonstrated that carvedilol has antioxidative properties by three independent approaches; lipid peroxidation, protein oxidation, and superoxide production. This property was significantly stronger than that of [alpha]-tocopherol. This is the first report to demonstrate the inhibitory effect of carvedilol on protein oxidation in the brain. Carvedilol may act as a neuroprotective agent in cerebrovascular disorders.

REFERENCES

1 Pellegrini-Giampietro DE, Cherici G, Alesiani M, Carla V, Moroni F. Excitatory amino acid release and free radical formation may cooperate in the genesis of ischemia-induced neuronal damage. J Neurosci 1990; 10: 1035-1041

2 Goto S, Nakamura A, Radak Z, et al. Carbonylated proteins in aging and exercise: Immunoblot approaches. Mech Ageing Dev 1999; 107: 245-253

3 Butterfield DA, Kanski J. Brain protein oxidation in age-related neurodegenerative disorders that are associated with aggregated proteins. Mech Ageing Dev 2001; 122: 945-962

4 Van SD, Chen X, Fu J, et al. RAGE and amyloid-beta peptide neurotoxicity in Alzheimer's disease. Nature 1996; 382: 685-691

5 Yue TL, Cheng HY, Lysko PG, et al. Carvedilol, a new vasodilator and beta adrenoceptor antagonist, is an antioxidant and free radical scavenger. J Pharmacol Exp Ther 1992; 263: 92-98

6 Yue TL, McKenna PJ, Lysko PG, Ruffolo RR Jr, Feuerstein GZ. Carvedilol, a new antihypertensive, prevents oxidation of human low density lipoprotein by macrophages and copper. Atherosclerosis 1992; 97: 209-216

7 Yue TL, McKenna PJ, Ruffolo RR Jr, Feuerstein G. Carvedilol, a new beta-adrenoceptor antagonist and vasodilator antihypertensive drug, inhibits Superoxide release from human neutrophils. Eur J Pharmacol 1992; 214: 277-280

8 Yue TL, McKenna PJ, Gu JL, Cheng HY, Ruffolo RR Jr, Feuerstein GZ. Carvedilol, a new antihypertensive agent, prevents lipid peroxidation and oxidative injury to endothelial cells. Hypertension 1993; 22: 922-928

9 Feuerstein R, Yue TL. A potent antioxidant, SB209995, inhibits oxygen-radical-mediated lipid peroxidation and cycotoxicity. Pharmacology 1994; 48: 385-391

10 Abreu RM, Santos DJ, Moreno AJ. Effects of carvedilol and its analog BM-910228 on mitochondrial function and oxidative stress. J Pharmacol Exp Ther 2000; 295: 1022-1030

11 Dandona P, Karne R, Ghanim H, Hamouda W, Aljada A, Magsino CH Jr. Carvedilol inhibits reactive oxygen species generation by leukocytes and oxidative damage to amino acids. Circulation 2000; 101: 122-124

12 Oettl K, Greilberger J, Zangger K, Haslinger E, Reibnegger G, Jurgens G. Radical-scavenging and iron-chelating properties of carvedilol, an antihypertensive drug with antioxidative activity. Biochem Pharmacol 2001; 62: 241-248

13 Nishida A, Kimura H, Nakano M, Goto T. A sensitive and specific chemiluminescence method for estimating the ability of human granulocytes and monocytes to generate O2. Clin Chim Acta 1989; 179: 177-181

14 Tanaka M, Sotomatsu A, Yoshida T, Hirai S, Nishida A. Detection of Superoxide production by activated microglia using a sensitive and specific chemiluminescence assay and microglia-mediated PC12h cell death. J Neurochem 1994; 63: 266-270

15 Yoshida T, Sotomatsu A, Tanaka M, Hirai S. Inhibitory effect of bifemelane on Superoxide generation by activated neutrophils measured using a simple chemiluminescence method. Free Radic Res 1994; 21: 371-376

16 Helliwell B, Gutteridge JM. Role of free radicals and catalytic metal ions in human disease: An overview. Methods Enzymol 1990; 186: 1-85

17 Stadtman ER. Oxidation of free amino acids and amino acid residues in proteins by radiolysis and by metal-catalyzed reactions. Annu Rev Biochem 1993; 62: 797-821

18 Levine RL, Oliver CN, Fulks RM, Stadtman ER. Turnover of bacterial glutamine synthetase: Oxidative inactivation precedes proteolysis. Proc Natl Acad Sci USA 1981; 78: 2120-2124

19 Rivett AJ. Preferential degradation of the oxidatively modified form of glutamine synthetase by intracellular mammalian proteases. J Biol Chem 1985; 260: 300-305

20 Wolff SP, Dean RT. Fragmentation of proteins by free radicals and its effect on their susceptibility to enzymic hydrolysis. Biochem J 1986; 234: 399-403

21 Davies KJ, Delsignore ME. Protein damage and degradation by oxygen radicals. III. Modification of secondary and tertiary structure. J Biol Chem 1987; 262: 9908-9913

22 Shiga Y, Onodera H, Kogure K, et al. Neutrophil as a mediator of ischemic edema formation in the brain. Neurosci Lett 1991; 125: 110-112

23 Zini I, Tomasi A, Grimaldi R, Vannini V, Agnati LF. Detection of free radicals during brain ischemia and reperfusion by spin trapping and microdialysis. Neurosci Lett 1992; 138: 279-282

Yoko Suzuki, Makoto Tanaka, Makoto Sohmiya, Toshihiko Yoshida and Koichi Okamoto

Department of Neurology, Gunma University Graduate School of Medicine, Gunma, Japan

Correspondence and reprint requests to: Makoto Tanaka, Department of Neurology, Gunma University School of Medicine, 3-39-22 Showamachi, Maebashi, Gunma 371-8511, Japan. [tanakama@showa.gunma-u.ac.jp] Accepted for publication April 2003.

Copyright Forefront Publishing Group Oct 2003
Provided by ProQuest Information and Learning Company. All rights Reserved

Return to Carvedilol
Home Contact Resources Exchange Links ebay