Você está na página 1de 31

Antihypertensives 1

Antihypertensives
Alexander Scriabine, Miles Laboratories, Inc., Institute for Preclinical Pharmacology, New Haven,
Connecticut 06511, United States
David G. Taylor, Miles Laboratories, Inc., Institute for Preclinical Pharmacology, New Haven, Connecticut
06511, United States

1. Introduction . . . . . . . . . . . . . . 1 4.2.1.1. Reserpine . . . . . . . . . . . . . . . . 11


2. Diuretics . . . . . . . . . . . . . . . . 2 4.2.1.2. Guanethidine . . . . . . . . . . . . . . 12
2.1. Sulfonamide Diuretics . . . . . . . 2 4.2.2. α-Adrenoceptor Antagonists . . . . 12
2.1.1. Hydrochlorothiazide . . . . . . . . . 3 4.2.2.1. Prazosin . . . . . . . . . . . . . . . . . 12
2.1.2. Chlorthalidone . . . . . . . . . . . . . 3 4.2.2.2. Indoramin . . . . . . . . . . . . . . . . 13
2.1.3. Furosemide . . . . . . . . . . . . . . . 3 4.2.2.3. Urapidil . . . . . . . . . . . . . . . . . 13
2.1.4. Mefruside . . . . . . . . . . . . . . . . 4 4.2.3. β-Adrenoceptor Antagonists . . . . 14
2.1.5. Indapamide . . . . . . . . . . . . . . . 4 4.2.3.1. Propranolol . . . . . . . . . . . . . . . 14
2.2. K+ -Retaining Diuretics . . . . . . . 4 4.2.3.2. Metoprolol . . . . . . . . . . . . . . . 15
2.2.1. Amiloride . . . . . . . . . . . . . . . . 4 4.2.3.3. Nadolol . . . . . . . . . . . . . . . . . 15
2.2.2. Triamterene . . . . . . . . . . . . . . . 5 4.2.3.4. Pindolol . . . . . . . . . . . . . . . . . 15
2.3. Other Diuretics . . . . . . . . . . . . 5 4.2.3.5. Timolol . . . . . . . . . . . . . . . . . . 16
2.3.1. Ethacrynic Acid . . . . . . . . . . . . 5 4.2.3.6. Acebutolol . . . . . . . . . . . . . . . . 16
2.3.2. Indacrinone . . . . . . . . . . . . . . . 6 4.2.3.7. Labetalol . . . . . . . . . . . . . . . . . 17
2.3.3. Muzolimine . . . . . . . . . . . . . . . 6
5. Smooth Muscle Relaxants . . . . . 17
3. Inhibitors of Angiotensin Con-
5.1. Ca2+ Channel Antagonists . . . . 17
verting Enzyme . . . . . . . . . . . . 6
5.1.1. Nifedipine . . . . . . . . . . . . . . . . 17
3.1. Captopril . . . . . . . . . . . . . . . . 7
5.1.2. Nitrendipine . . . . . . . . . . . . . . . 18
3.2. Enalapril . . . . . . . . . . . . . . . . 7
4. Inhibitors of the 5.1.3. Verapamil . . . . . . . . . . . . . . . . 19
Sympathetic Nervous System . . . 8 5.1.4. Diltiazem . . . . . . . . . . . . . . . . 19
4.1. Drugs with Primarily Central Ac- 5.2. Vasodilators with Other Mecha-
tions . . . . . . . . . . . . . . . . . . . 8 nisms of Action . . . . . . . . . . . . 20
4.1.1. Methyldopa . . . . . . . . . . . . . . . 9 5.2.1. Hydralazine . . . . . . . . . . . . . . . 20
4.1.2. Clonidine . . . . . . . . . . . . . . . . 9 5.2.2. Diazoxide . . . . . . . . . . . . . . . . 21
4.1.3. Guanfacine . . . . . . . . . . . . . . . 9 5.2.3. Minoxidil . . . . . . . . . . . . . . . . 21
4.1.4. Guanabenz . . . . . . . . . . . . . . . 10 5.2.4. Sodium Nitroprusside . . . . . . . . . 21
4.2. Drugs with Primarily Peripheral 6. Perspectives in the Development of
Action . . . . . . . . . . . . . . . . . . 10 Antihypertensive Drugs . . . . . . 22
4.2.1. Amine Depleting and Adrenergic 7. Summary and Conclusions . . . . 22
Neuron Blocking Agents . . . . . . . 11 8. References . . . . . . . . . . . . . . . 23

1. Introduction The available antihypertensive drugs can be


classified on the basis of their mechanism of ac-
In spite of the fact that many drugs are now avail- tion as follows: (1) diuretics, (2) inhibitors of an-
able for the treatment of hypertension, new drugs giotensin converting enzyme (ACE), (3) drugs
are being introduced every year. The existence of which interfere with the sympathetic nervous
a lucrative and still expanding market as well as system, and (4) vascular smooth muscle relax-
the medical need for effective antihypertensive ants. This article describes the major represen-
drugs with minimal side effects stimulate con- tatives of each of the above classes of antihyper-
tinuous research activity in this field. tensive drugs.

c 2005 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim


10.1002/14356007.a04 235
2 Antihypertensives

2. Diuretics tivity and to reduce the side effects of sulfa-


nilamide led to the development of acetazol-
The chemistry, pharmacology, and clinical use amide [59-66-5] [12], which can be considered
of diuretics in hypertension and edema are dis- the prototype of sulfonamide diuretics. Its con-
cussed in [1], also → Diuretics. The rapid ex- tinuous use led to metabolic acidosis and to the
pansion of the diuretic field occurred after the development of tolerance to its diuretic effect.
introduction of acetazolamide [2] and chlorothi- The search for a better diuretic led to the devel-
azide [3], [4]. The discovery of the effectiveness opment of chlorothiazide [58-94-6] [13]. The
of chlorothiazide in the therapy of hypertension diuretic activity of chlorothiazide, unlike that
[5] greatly expanded the market for diuretics and of acetazolamide, is not determined by its car-
led to the synthesis of many chemically related bonic anhydrase inhibitory activity. At therapeu-
compounds. Since hypokalemia is one of the ma- tic doses, chlorothiazide increases the excretion
jor side effects of chlorothiazide and related thi- of chloride rather than that of hydrogen carbon-
azides, attempts were made to develop diuretics ate. By repeated administration chlorothiazide
with K+ -retaining or isokalemic properties. Al- produces effective diuresis without metabolic
dosterone antagonists, such as spironolactone or acidosis.
other K+ retaining diuretics, e.g., amiloride [6] Extensive structure – activity relationship
or triamterene [7] were shown to produce diure- studies led subsequently to the development of
sis while reducing excretion of potassium. Com- chemically and pharmacologically related thia-
binations of thiazides with potassium retaining zide diuretics which differed from chlorothia-
diuretics (e.g., Moduretic or Dyazide) have di- zide either in the relative potency or in the dura-
uretic effects and lower arterial pressure without tion of action. The most commonly used thiazide
any change in the excretion of potassium. These diuretic is hydrochlorothiazide [14], which is ca.
combinations are widely used in general prac- 10 times more potent than chlorothiazide. The
tice. most potent thiazide among marketed drugs is
The mechanism of antihypertensive action cyclopenthiazide [15], while methylclothiazide
of diuretics is still a subject of controversy, al- [16] and polythiazide [17] have exceptionally
though numerous studies suggested that the na- long durations of action. Their longer duration
triuretic action of diuretics is primarily responsi- of action is due to higher lipid solubility (and
ble for their antihypertensive effect. The kidneys therefore greater reabsorption of the drug in the
of hypertensive animals or humans can excrete proximal tubules) and higher protein binding.
salt in normal amounts but only at elevated ar- Other clinically important sulfonamide diuretics
terial pressure levels and diuretics increase the include mefruside [18], [19], indapamide [20],
excretion of salt independent of blood pressure and chlorthalidone [21].
level [8]. Also the increase in sodium in the vas- A significant advance in diuretic therapy was
cular smooth muscle cell was correlated with the the introduction of furosemide [12], which has
increase in the intracellular calcium, and there- much higher ceiling natriuretic and chloruretic
fore, with the vasoconstrictor tone leading to the effects than thiazide diuretics. The high ceiling
elevation of arterial pressure [9], [10]. The na- effect of furosemide is determined by the site of
triuretic effect of diuretics tends to prevent ex- its tubular action: furosemide inhibits chloride
cessive accumulation of sodium. and sodium reabsorption in the ascending limb
of Henle’s loop (“loop” diuretic). Furosemide is
widely used clinically, although in hypertension
2.1. Sulfonamide Diuretics it is not more effective than thiazide diuretics.
An attempt to find a “loop” diuretic with a long
The first sulfonamide shown to have diuretic ac- duration of action led to the development of mu-
tivity in humans was sulfanilamide [63-74-1] zolimine [21], which is not yet available on the
[11]. Subsequent attempts to improve the ac- United States market.
Antihypertensives 3

2.1.1. Hydrochlorothiazide acting than hydrochlorothiazide [21]. The du-


ration of action is determined by its elimina-
tion half-life (t 1/2 ) of 49 h in humans. Be-
cause of longer duration of action, chlorthali-
done can be given every 72 h. Chlorthalidone
is widely used in the treatment of hyperten-
sion: it can be used alone or in combination
with methyldopa or β-adrenoceptor antagonists.
Hydrochlorothiazide [58-93-5], 6-chloro- Side effects are also similar to those of thiazides
3,4-dihydro-2H-1, 2, 4-benzothiadiazine-7-sul- and include hypokalemia, hyperuricemia, hy-
fonamide 1,1-dioxide, C7 H8 ClN3 O4 S2 , M r perglycemia, and hyperlipoproteinemia (eleva-
297.72, mp 273 – 275 ◦ C, is insoluble in water, tion of low-density lipoproteins). Clinical doses
soluble in NaOH, ethanol, and acetone. Hydro- range from 25 to 400 mg.
chlorothiazide is widely used in the treatment
of hypertension or edema.
Trade names: HydroDiuril, Esidrix. 2.1.3. Furosemide
Like other benzothiadiazine-type diuretics
(or thiazides), hydrochlorothiazide increases the
excretion of sodium and chloride independent
of acid – base balance. Hydrochlorothiazide also
increases the excretion of potassium. The result-
ing hypokalemia represents its major side ef-
fect. Other side effects include hyperuricemia,
hyperglycemia, and hyperlipoproteinemia. The Furosemide [54-31-9], 5-(aminosulfonyl)-
relative diuretic potency of hydrochlorothiazide 4-chloro-2-[(2-furanylmethyl)amino]benzoic
is ca. 10 times higher than that of chlorothia- acid, 4-chloro-N-furfuryl-5-sulfamoylanthran-
zide [14]. The efficacy (ceiling effect) is similar ilic acid, C12 H11 ClN2 O5 S, M r 330.77, mp
to that of other thiazide diuretics and is con- 206 ◦ C, is only slightly soluble in water, sol-
siderably lower than that of furosemide. Daily uble in methanol, acetone, and dilute NaOH.
clinical doses range from 25 to 200 mg and are Trade name: Lasix.
given with potassium supplements or in combi- The major site of action of furosemide is
nation with potassium-retaining diuretics to pre- in the ascending limb of Henle’s loop, prox-
vent hypokalemia. imal to the thiazide diuretics. Like thiazides,
furosemide increases the excretion of sodium,
chloride, and potassium, but its efficacy (ceil-
2.1.2. Chlorthalidone ing effect) is much higher than that of thia-
zides and similar diuretics [12]. The duration
of diuretic action of furosemide is short and is
correlated with its elimination half-life (0.85 h).
Independent of its diuretic action, furosemide,
by i.v. administration, tends to increase venous
capacitance and to reduce left ventricular end
diastolic pressure. In spite of its greater di-
Chlorthalidone [77-36-1], 2-chloro-5-(1- uretic efficacy, furosemide is not more effective
hydroxy-3-oxo-1-isoindolinyl)benzenesulfon- than thiazide diuretics in the treatment of hy-
amide, C14 H11 ClN2 O4 S, M r 338.78, mp pertension. Side effects of furosemide include
224 – 226 ◦ C, is slightly soluble in water those of thiazide diuretics: hypokalemia, hy-
(12 mg/100 mL), more soluble in diluted NaOH perglycemia, hyperuricemia, and hyperlipopro-
or warm ethanol. teinemia. In addition, furosemide tends to pro-
Trade name: Hygroton. duce metabolic alkalosis and ototoxicity, which
Chlorthalidone is pharmacologically similar is typical for “loop” diuretics. Daily clinical
to thiazide diuretics but is substantially longer doses of furosemide range from 20 to 200 mg.
4 Antihypertensives

2.1.4. Mefruside Side effects of indapamide include hypo-


kalemia, hyperuricemia, and metabolic alkalo-
sis, and are usually observed at doses higher
than required for satisfactory control of arterial
pressure in hypertensive patients (5 mg/d and
higher). The safe and effective clinical dose is
2.5 mg/d.
Mefruside [7195-27-9], 4-chloro-N 1 -meth-
yl-N 1 -[(tetrahydro-2-methyl-2-furanyl)meth-
yl]-1,3-benzenedisulfonamide, 4-chloro-N 1 - 2.2. K+ -Retaining Diuretics
methyl-N 1 -(tetrahydro-2-methylfurfuryl)-m- The development of K+ -retaining (or sparing)
benzenedisulfonamide, C13 H19 ClN2 O5 S2 , M r diuretics was stimulated by the fact that K+ loss
382.90, mp 149 – 150 ◦ C (dl-crystals), 146 ◦ C and consequent hypokalemia represent the ma-
(d-form). The d-form is more active as a diuretic jor side effect of sulfonamide diuretics. It was
than the l-form. considered possible to develop K+ -retaining di-
Trade name: Baycaron. uretics because aldosterone was known to retain
The primary site of action of mefruside is the sodium while increasing the excretion of potas-
cortical diluting segment; duration of action is sium. The search for aldosterone inhibitors led
20 to 24 h. Mefruside has carbonic anhydrase in- to the development of spironolactone [52-01-7]
hibitory activity but is less potent than acetazol- [22], which is still used clinically. The activity
amide [18], [19]. One of its metabolites, oxome- of spironolactone is determined by its ability to
fruside, also has diuretic activity. Side effects of compete with aldosterone at its receptor sites.
mefruside are similar to those of thiazide diuret- It is inactive in the absence of aldosterone but
ics, in that it produces hyperuricemia and hypo- has diuretic, natriuretic, and K+ retaining effects
kalemia. Daily clinical doses range from 25 to in its presence. The pharmacological effects of
200 mg. In hypertension, mefruside is effective spironolactone are to a great extent due to its
at 25 – 50 mg/d or every other day. active metabolite, canrenone, which is formed
within 3 min after administration of spironolac-
tone and has a half-life of ca. 12 h.
2.1.5. Indapamide
The clinical use of spironolactone is severely
limited by side effects, including hyperkalemia,
gynecomastia, impotence, and menstrual cycle
abnormalities. In rats chronic spironolactone
treatment was tumorigenic, and therefore, its
use is restricted. Because of these side effects
and the high cost of spironolactone, the search
Indapamide [26807-65-8], 3-(amino- for nonsteroidal aldosterone inhibitors contin-
sulfonyl)-4-chloro-N-(2,3-dihydro-2-meth- ued and led to the development of K+ -retaining
yl-1H-indol-1-yl)benzamide, 4-chloro-N-(2- diuretics that increase sodium excretion without
methyl-1-indolinyl)-3-sulfamoylbenzamide, inhibiting aldosterone.
C16 H16 ClN3 O3 S, M r 365.84, mp 160 – 162 ◦ C, Two major K+ -retaining diuretics which do
pK a 8.3, is lipid soluble. not inhibit aldosterone are amiloride and tri-
Trade names: Fludex, Ipamox, Natrilix. amterene.
The site of diuretic action of indapamide is
similar to that of thiazide diuretics: the prox- 2.2.1. Amiloride
imal part of distal tubules. Its duration of di-
uretic action can exceed 24 h. Indapamidediffers
from thiazide diuretics in having additional di-
rect smooth muscle relaxant effect and hypoten-
sive effect at doses below those required to pro-
duce diuresis [20].
Antihypertensives 5

Amiloride [2609-46-3], 3,5-diamino-N- uretic effect of hydrochlorothiazide is antago-


(aminoiminomethyl)-6-chloropyrazinecarbox- nized by triamterene.
amide, N-amidino-3,5-diamino-6-chloropyra- The major side effect of triamterene alone is
zinecarboxamide, C6 H8 ClN7 O, M r 229.65, mp hyperkalemia. Nausea, leg cramps, and dizzi-
293.5 ◦ C, is soluble in water. It is widely used ness have also been described. The initial clini-
in combination with hydrochlorothiazide (Mod- cal dose is 100 mg twice daily. Dyazide capsules
uretic). contain 50 mg of triamterene and 25 mg of hy-
Trade names: Colectril and Midamor. drochlorothiazide and are given 1 – 4 times daily.
The site of action of amiloride is in the dis-
tal portion of distal tubules and in the collecting
duct, where amiloride inhibits sodium reabsorp- 2.3. Other Diuretics
tion and potassium secretion [6]. It is thought
to block the Na+ channel and the Na+ /H+ ex- In addition to sulfonamides and pyrazines, di-
change pump. In addition to the renal tubules, uretic activity has been discovered among many
amiloride blocks Na+ channels in various ep- other chemical classes of drugs. Among them
ithelial membranes, including the salivary ducts. are aryloxyacetic acids, 2-aminomethylphenols,
In hypertension, if used alone, amiloride is not as and various heterocyclic compounds. Because
effective as hydrochlorothiazide. The major side all diuretics cannot be covered in this article,
effect of amiloride is hyperkalemia. Other side compounds that are important therapeutically
effects include nausea, anorexia, and abdominal as well as historically have been selected. The
pain. three diuretics discussed (ethacrynic acid, in-
The clinical dose range of amiloride is dacrinone, and muzolimine) differ from sulfon-
5 – 20 mg/d. Potassium supplements are contra- amides and pyrazines in their pharmacological
indicated with amiloride; other K+ -retaining di- profiles and represent three different chemical
uretics should not be combined with amiloride. classes.

2.2.2. Triamterene 2.3.1. Ethacrynic Acid

Ethacrynic acid [58-54-8], [2,3-dichloro-4-


Triamterene [396-01-0], 6-phenyl-2,4,7- (2-methylene-1-oxybutyl)phenoxy]acetic acid,
pteridinetriamine, 2,4,7-triamino-6-phenyl- C13 H12 Cl2 O4 , M r 303.14, mp 121 – 125 ◦ C, is
pteridine, C12 H11 N7 , M r 253.26, mp 316 ◦ C, only slightly soluble in water but very soluble in
is insoluble in water or chloroform and only alcohol (1 g/1.6 mL).
slightly soluble in alcohol. The efficacy (ceiling effect) of ethacrynic
Trade names: Dyrenium; Dyazide (combi- acid is similar to that of furosemide and con-
nation with hydrochlorothiazide). siderably higher than that of hydrochlorothia-
Triamterene increases urinary excretion of zide. Ethacrynic acid increases the excretion of
sodium and chloride and decreases excretion of sodium and chloride. It also slightly increases
potassium and hydrogen ions. The primary site the excretion of potassium. The major site of
of triamterene action is in the distal tubules [7]. action is the ascending limb of Henle’s loop
The biochemical effects of triamterene are not [13]. The biochemical mechanism of action is
well-known, although triamterene is capable of not known precisely, although at high concen-
forming “false” nucleotides. trations ethacrynic acid inhibits renal Na+ −K+
The principal use of triamterene is in combi- dependent ATPase and oxygen consumption in
nation with hydrochlorothiazide. The natriuretic medullary tubules. Ethacrynic acid reacts with
effects of both diuretics are additive, but the kali- thiols in the body fluids and forms adducts which
are likely to represent the active forms of this
6 Antihypertensives

drug. The side effects of ethacrynic acid include 2.3.3. Muzolimine


abdominal discomfort, nausea, hyperuricemia,
neutropenia, thrombocytopenia, ototoxicity, and
metabolic alkalosis. The usual clinical doses are
50 mg twice a day; the maximal daily dose is
400 mg.

Muzolimine [55294-15-0], 5-amino-2-[1-


2.3.2. Indacrinone (3,4-dichlorophenyl)-ethyl]-2,4-dihydro-3H-
pyrazol-3-one, C11 H11 Cl2 N3 O, M r 272.13, mp
127 –129 ◦ C, is insoluble in water, but is solu-
ble in ethanol, polyethylene glycol, or dimethyl
sulfoxide. It is highly lipophilic, with a pK a
value of 9.3 and a partition coefficient in oc-
tanol/water of 2.29.
Trade name: Edrul.
The site of diuretic action of muzolimine is
the ascending limb of Henle’s loop. Unlike other
loop diuretics muzolimine does not act at the lu-
minal site. It appears to inhibit sodium and chlo-
ride transport at the peritubular site of the cell
membrane. Muzolimine exhibits a high-efficacy
(ceiling) effect similar to that of furosemide, but
Indacrinone [57296-63-6], [(6,7-dichloro- has a considerably longer duration of action. The
2,3-dihydro-2-methyl-1-oxo-2-phenyl-1H- duration of antihypertensive action of muzolim-
inden-5-yl)oxy]acetic acid, M r 365.19. A 9 : 1 ine at a single dose exceeds 24 h [26], [27].
ratio of (+)- to (−)-indacrinone enantiomers was Clinical doses of muzolimine range from 10
studied as MK-286 and found to be isouricemic to 480 mg; good therapeutic results were ob-
during chronic use. The (+)-enantiomer is urico- tained with doses of 30 mg/d in most patients.
suric, whereas the (−)-enantiomer retains uric Muzolimine is particularly useful in the treat-
acid. The racemic mixture was tested as MK-196 ment of edema associated with renal failure.
[23–25].
Indacrinone and its enantiomers lower arte-
rial pressure in hypertensive animals and in hu-
mans, inhibit sodium and chloride reabsorption
3. Inhibitors of Angiotensin
in the ascending limb of Henle’s loop, and in- Converting Enzyme
crease excretion of potassium. Hypokalemia can
be antagonized by amiloride. The site of action The renin – angiotensin system is involved in
of enantiomers in the renal tubules appears to the control of blood pressure in normotensive
differ. The (−)-enantiomer has its primary action and hypertensive states. The conversion of an-
in the medullary portion of the ascending limb, giotensinogen to angiotensin II involves a two-
while the (+)-enantiomer has its primary action step sequence:
in the cortical diluting segment. The saluretic
efficacy of indacrinone is similar to or greater
than that of furosemide, while the duration of
action is longer. Side effects of indacrinone in
humans include headache, lethargy, weakness,
dry mouth, and slight, but transient elevations in
SGOT and/or SGPT. The clinical doses of the
(−)-enantiomer ranged from 10 to 40 mg, while
the (+)-enantiomer was given at doses from 10
to 400 mg.
Antihypertensives 7

Pharmacological approaches for interference The hypotensive responses are not usually ac-
with the renin – angiotensin system have fo- companied by a reflex increase in heart rate. Cap-
cused on renin inhibitors, angiotensin con- topril facilitates the reflex slowing of heart rate
verting enzyme (ACE) inhibitors, and an- in some manner [38]. This could involve the re-
giotensin II receptor antagonists [28]. Captopril moval of facilitatory effects of angiotensin II on
was the first orally effective ACE inhibitor ap- sympathetic nervous transmission to the heart
proved for use in mild-to-moderate hyperten- or a central inhibitory effect of angiotensin II on
sion. Enalapril was developed later as a nonsulf- cardiac vagal tone [49]. Further studies have also
hydryl-containing inhibitor. indicated that captopril possesses cardioprotec-
tive properties [50].
Captopril decreases renal vascular resistance,
3.1. Captopril and increases sodium and water excretion [51],
[52].
Renin levels are increased and aldosterone
levels are reduced by captopril. A certain num-
ber of patients with essential hypertension ex-
hibit a subnormal aldosterone secretory re-
sponse to angiotensin II challenge [53]. The ab-
Captopril [62571-86-2], 1-(3-mercapto-2- normal responders also exhibit higher baseline
methyl-1-oxopropyl)-l-proline, C9 H15 NO3 S, plasma angiotensin II and lower levels of aldos-
M r 217.28, mp 87 – 88 ◦ C, is soluble in water, terone. Captopril treatment for 72 h removed the
alcohol, and chloroform; difference in baseline values and partially cor-
Trade name: Capoten. rected the abnormal aldosterone response to an-
The rationale and design of the angiotensin giotensin II.
converting enzyme (ACE) inhibitors have been The absorption of captopril in humans is rapid
described [29], [30]. The effectiveness of cap- and extensive; 75 % of the radioactivity after
topril on ACE was shown by its ability to in- a single oral dose of 14 C-captopril was recov-
hibit the rise in blood pressure induced by an- ered in the urine within 48 h. The metabolites
giotensin I, while leaving unchanged the re- of captopril include disulfides and endogenous
sponses to angiotensin II [31]. sulfhydryl compounds, e.g., cysteine.
It has been postulated that the antihyper- The side effects of captopril include protein-
tensive mechanism may include not only in- uria, maculopapular rash, itch, disturbance of
hibition of plasma ACE activity, but also in- taste, and occasional leukopenia [54].
hibition of local tissue ACE, especially in the
vascular smooth muscle [32–35]. In addition,
a number of alternative mechanisms could ex- 3.2. Enalapril
plain the efficacy of ACE inhibitors in nonrenin-
dependent hypertension [36]. The alternatives
include (1) interference with α-adrenergic vaso-
constriction [37–42]; (2) a central inhibition of
sympathetic nervous outflow [35], [43]; (3) el-
evation in bradykinin or prostaglandins, due to
the similarity of kininase II and ACE [31], [32], Enalapril [75847-73-3], 1-[N-[1-(ethoxy-
[44]; and (4) alteration in the permeability of carbonyl)-3-phenylpropyl]-l-alanyl]-l-proline,
vascular smooth muscle to sodium ions [45]. C20 H28 N2 O5 , M r 376.45, mp 143 – 144.5 ◦ C,
Clinically, the antihypertensive actions of is a white to off-white crystalline powder, sol-
captopril are associated with a reduction in total uble in methanol and slightly soluble in water.
peripheral resistance and a slight to moderate in- Unlike captopril, enalapril does not contain the
crease in cardiac output [46], [47]. In malignant sulfhydryl moiety which has been implicated in
hypertensive patients captopril was very effec- some adverse side effects, such as rash and loss
tive during acute and long-term treatment [48]. of taste.
8 Antihypertensives

Enalapril is readily absorbed by oral admin- was reduced insignificantly [62]. In normoten-
istration while the active diacid form is poorly sive subjects, the direction of change in nor-
absorbed. Deesterification of the prodrug takes epinephrine levels was dependent on the sodium
place primarily in the liver [55]. Enalapril is intake [66], [67].
about 17-fold more potent than captopril for in- The mechanism of action of enalapril remains
hibition of hog plasma ACE. In both rats and to be fully elucidated. The dominant mechanism
dogs, enalapril inhibits the pressor responses to most certainly involves the renin –angiotensin
angiotensin I for approximately 6 h. Although system; however, additional mechanisms must
the onset of action is somewhat slower, the po- also be considered.
tency of enalapril in vivo is about 4 – 8 fold The major toxicological findings in dogs
greater than that of captopril in the same species were renal functional changes, and renal tubu-
[56]. lar degeneration. Toxic actions of enalapril are
Similar to captopril, the antihypertensive ac- exacerbated by coadministration of hydrochlo-
tion of enalapril is most pronounced in high- rothiazide and attenuated by saline supplements
renin forms of hypertension and enalapril also [55]. In humans the major side effects include
exhibits antihypertensive activity in low-renin rash, angioedema, proteinuria, and renal insuf-
models [57]. ficiency [54].
In clinical studies, enalapril effectively low-
ers arterial pressure when given as sole therapy
or in combination with diuretics [58–60]. Simi- 4. Inhibitors of the Sympathetic
lar to preclinical studies, the onset of inhibition
of ACE in humans is slower than with capto-
Nervous System
pril. This finding has been attributed to the time
needed for the deesterification reaction [61]. 4.1. Drugs with Primarily Central
Reflex tachycardia is seldom observed with Actions
enalapril [62], [63]. Cardiac function is well-
maintained [62], [64] and, in one group of hy- Some of the widely used antihypertensive drugs
pertensive patients, the left ventricular mass was are thought to lower arterial pressure by centrally
reduced during therapy [62]. induced blockade of sympathetic tone and con-
With regard to renal function, enalapril in- sequent reduction of peripheral vascular resis-
creases sodium excretion, glomerular filtration tance. The central control of the sympathetic ner-
rate, and renal blood flow in sodium-restricted vous system is mediated by various neurotrans-
dogs [55]. In spontaneously hypertensive rats mitters, including norepinephrine, and by drugs
(SHR), antihypertensive doses of enalapril do which enhance norepinephrine release or stim-
not cause sodium and water retention upon ulate postsynaptic α-adrenoceptors involved in
chronic administration [65]. Renal vasodilata- the regulation of sympathetic tone. The 5HT
tion and natriuresis have been observed in hy- agonists, dopaminergic, or GABA ergic drugs,
pertensive patients treated with enalapril [62], were also shown or are assumed to control hy-
[63]. pertension by a central action. It is presently not
Hormonal responses during acute and clear whether their effects are mediated through
chronic (3 months) enalapril treatment consist central inhibition of the sympathetic nervous
of decreased angiotensin II and aldosterone lev- system or by other central mechanisms.
els and a rise in plasma renin activity. In addi- The major goal in the development of new
tion, bradykinin and PGE2 were unchanged by a centrally acting drugs is to achieve a high degree
single dose of enalapril [63], [66]. Epinephrine of selectivity for blood pressure control, which
levels were consistently decreased by enalapril, would lead to antihypertensive action without
whereas, in hypertensive patients during chronic sedative, skeletal muscle relaxant, or emetic side
therapy with enalapril, plasma norepinephrine effects. For further details on the central control
of arterial pressure by drugs see reviews [68],
[69].
Antihypertensives 9

4.1.1. Methyldopa ca. 300 ◦ C, pK a 8.2, is easily soluble in wa-


ter (1 g/13 mL) and ethanol (1 g/25 mL).
Trade names: Catapres, Catapressan.
Clonidine is an α-adrenoceptor stimu-
lant, which stimulates preferentially α2 -adren-
oceptors [81], [82]. The stimulation of central
α-adrenoceptors by clonidine leads to inhibition
The antihypertensive activity of methyldopa
of central sympathetic tone, resulting in a lower-
[555-30-6], 3-hydroxy-α-methyl-l-tyrosine,
ing of arterial pressure and of heart rate [83–87].
C10 H13 NO4 , M r 211.21, was first attributed
Clonidine has many other pharmacological
to the inhibition of aromatic amino acid de-
effects. The most prominent central effect of
carboxylase [70] and later to a formation of a
clonidine is sedation. Clonidine decreases spon-
“false transmitter” [71], [72]. It is postulated that
taneous motor activity and produces ataxia and
α-methylnorepinephrine, a methyldopa metabo-
catatonia as well as signs of increased periph-
lite, lowers arterial pressure by stimulating cen-
eral sympathetic activity in animals, i.e., pilo-
tral adrenoceptors, which causes a reduction in
erection and exophthalmus [88]. Clonidine has
sympathetic nervous outflow [73], [74].
antinociceptive activity in mice [89] and is ef-
The antihypertensive effect of methyldopa in
fective in the management of withdrawal reac-
humans is associated with a reduction in the pe-
tion in opiate addicts [90]. Intraocular pressure is
ripheral vascular resistance and in the plasma
decreased by clonidine [91]. Clonidine reduces
renin activity [74–76]. Methyldopa increases
salivary secretion and has a biphasic effect on
coronary and cerebral blood flow [77], [78].
gastric secretion: inhibition at lower and stimu-
The oral absorption of methyldopa in humans
lation at higher doses [88], [92]. In rats and dogs
was reported to vary from 26 to 74 % [79].
clonidine has a diuretic effect which appears to
The peak plasma levels of methyldopa are ob-
be mediated by inhibition of ADH release [93].
served 3 – 6 h after oral dose. The major urinary
Clonidine is almost completely absorbed by
metabolites of methyldopa are methyldopa-o-
oral administration; its bioavailability in hu-
sulfate and 3-methoxymethyldopa. About 80 to
mans is ca. 75 % and the drug has no extensive
90 % of the oral dose of methyldopa is elimi-
first-pass metabolism [94]. In humans ca. 50 %
nated within 48 h after treatment [80].
of clonidine is excreted unchanged. The major
The initial dose of methyldopa in mild hyper-
metabolites of clonidine are p-hydroxyclonidine
tension is 250 mg t.i.d. (3 times/d), whereas in
and dichlorophenylguanidine [84].
severe hypertension methyldopa is used at doses
The major undesirable effect of clonidine is
up to 500 mg q.i.d. (4 times/d). The clinical ad-
the withdrawal reaction, which consists of a sud-
vantages of methyldopa include its effectiveness
den rise in arterial pressure, nervousness, agi-
in controlling hypertension, many years of clin-
tation, and an increase in heart rate [95], [96].
ical experience, and favorable hemodynamic ef-
Other side effects include dry mouth and seda-
fects, including maintenance of renal circulation
tion.
and decrease in plasma renin activity. Side ef-
The recommended starting dose of clonidine
fects of methyldopa include sedation, positive
is 0.1 mg/d, but it should not exceed a total daily
direct Coomb’s test, hepatotoxicity, and allergic
dose of 1 mg. The pharmacology of clonidine is
reactions.
reviewed extensively elsewhere [84].

4.1.2. Clonidine 4.1.3. Guanfacine

Clonidine [4205-90-7], 2-(2,6-dichloroanilino)- Guanfacine [29110-47-2], N-amidino-2-(2,6-


2-imidazoline, C9 H9 Cl2 N3 , M r 230.10, mp dichlorophenyl)acetamide, C9 H9 Cl2 N3 O, M r
10 Antihypertensives

246.08, mp 214 – 218 ◦ C, is water soluble with sistance with little or no effect on myocardial
a pK a of 7.1. Guanfacine is one of the most function.
potent members of a class of antihypertensive In animals or humans guanabenz has no ad-
phenylacylguanidines [97]. verse effects on renal function except a slight and
Trade names: Estulic, Estuline. transient fall in filtration rate after the first dose
Guanfacine is similar to clonidine in that [109], [110]. Under certain conditions guana-
the mechanism of antihypertensive action is at- benz increased excretion of water, and this effect
tributed largely to stimulation of central adreno- was attributed to the antagonism of vasopressin
ceptors [98], [99]. Guanfacine is claimed to dif- [111].
fer from clonidine in having less depressant ef- In humans guanabenz is well absorbed and
fect on the central nervous system (CNS) and exhibits a variable half-life of 7 – 10 h. Less
possibly may act at a different site within the than 1 % of the dose is excreted as unchanged
CNS [100], [101]. Like clonidine, guanfacine drug. The major metabolites of guanabenz in
exhibits a peripheral vasoconstrictor effect that rat, swine, Rhesus monkeys, and humans are p-
is mediated most likely by stimulation of α2 - hydroxyguanabenz and the glucuronide conju-
adrenoceptors. Guanfacine is rapidly absorbed gate [110], [112]. Clinical studies demonstrated
following oral administration with a bioavail- antihypertensive efficacy of guanabenz at doses
ability of 100 %. The half-life is approximately from 16 to 65 mg/d [113], [114]. The most com-
21 h after oral dosing. The major metabolite in mon side effects of guanabenz are dry mouth,
humans is the 3-hydroxy derivative of guan- sedation, weakness, and tiredness.
facine [100].
Dryness of mouth and sedation are the major
side effects of guanfacine in the clinic. The inci-
dence of side effects declines considerably with 4.2. Drugs with Primarily Peripheral
the duration of treatment. The withdrawal syn- Action
drome (rise in blood pressure and tachycardia)
was seen after abrupt discontinuation of guan- Drugs can inhibit sympathetic transmission at
facine in 2 – 4 % of the patients, but was less various peripheral sites: (1) sympathetic ganglia,
pronounced than after withdrawal of clonidine. (2) sympathetic nerve endings, or (3) postsynap-
The average dose of guanfacine in clinical trials tic adrenoceptors. Ganglionic blocking drugs
was 3 mg/d. The optimal effect without side ef- were used in the treatment of hypertension 20
fects was obtained at a single dose of 2 mg [100], to 30 years ago. Hexamethonium [60-26-4], a
[102], [103]. standard ganglionic blocking drug, is still used
today as a tool in experimental pharmacology.
Typical ganglionic blocking drugs (e.g., pen-
4.1.4. Guanabenz tolinium, chlorisondamine, mecamylamine, and
pempidine) block nicotinic receptors at the post-
synaptic membrane of the sympathetic ganglia
and prevent acetylcholine-induced depolariza-
tion. However, they are obsolete in the treat-
ment of hypertension because of unacceptable
side effects, which include atony of the gastroin-
Guanabenz [5051-62-7], 2-[(2,6-dichloro- testinal tract, inhibition of gastric and salivary
phenyl)methylene]hydrazinecarboximidamide, secretion, incomplete emptying of the urinary
[(2,6-dichlorobenzylidene)amino]guanidine, bladder, cycloplegia, and impotence. These side
C8 H8 Cl2 N4 , M r 231.07, mp 227 – 229 ◦ C. effects are to a great extent due to the blockade
Trade names: Rexitene, Wytensin. of parasympathetic ganglia. Blockade of sympa-
Like clonidine or guanfacine, guanabenz thetic ganglia can lead to additional side effects,
stimulates central α-adrenoceptors and lowers e.g., postural hypotension, syncope, and inhibi-
arterial pressure [104–108]. In hypertensive pa- tion of local vasomotor reflexes. Today the only
tients oral administration of guanabenz lowers indication remaining for ganglionic blockade is
arterial pressure and total peripheral vascular re- to produce so-called “controlled hypotension”
Antihypertensives 11

in surgery in order to reduce bleeding, facili- 4.2.1.1. Reserpine


tate small vessel surgery, and improve myocar-
dial performance by reducing preload and after-
load. For this indication, trimethaphan camsy-
late [68-91-7] (Arfonad) by infusion is the drug
of choice. Its short duration of action permits
precise control of blood pressure during infu-
sion and blood pressure returns to control levels
immediately after discontinuation of the infu-
sion.
The search for new ganglionic blocking drugs Reserpine [50-55-5], 11,17-dimethoxy-18-
was largely discontinued by 1960. It became [(3,4,5-trimethoxybenzoyl)oxy]yohimban-16-
clear that hypertension could be controlled by carboxylic acid methyl ester, C33 H40 N2 O9 , M r
drugs relatively free from the disabling effects 608.70, mp 264 – 265 ◦ C (decomp.), is poorly
accompanying ganglionic blockade. The drugs soluble in water, but is soluble in alcohol, glacial
which replaced the ganglionic blocking agents acetic acid, and chloroform. Reserpine was orig-
included diuretics and drugs which inhibited the inally isolated from roots of Rauwolfia ser-
sympathetic nervous system at loci peripheral to pentina L. Benth [115].
the ganglia; namely, at adrenergic nerve endings Synthesis: [116].
or at postsynaptic adrenoceptor sites. Trade names: Alserin, Reserpex, Serpasil.
Reserpine depletes the transmitters from
adrenergic, dopaminergic, and serotonergic neu-
4.2.1. Amine Depleting and Adrenergic rons. Depletion of norepinephrine from adren-
Neuron Blocking Agents ergic nerve endings is thought to be primarily
responsible for the antihypertensive action of re-
At the sympathetic nerve endings drugs can serpine. The onset of the antihypertensive action
decrease neurotransmitter (norepinephrine) re- is gradual. The maximal effect is seen 6 – 12 h
lease by depleting it from the storage sites in after a single dose of reserpine. The antihyper-
the nerve terminals or by decreasing the re- tensive effect is associated with a reduction in
lease by stimulation of inhibitory presynaptic cardiac output and in peripheral vascular resis-
α2 -adrenoceptors. tance. After repeated administration cardiac out-
Antihypertensive drugs which lower arterial put returns to control values, while peripheral
pressure primarily by an action at the sym- resistance remains at a reduced level [117–119].
pathetic nerve endings include reserpine and Reserpine is well absorbed by oral adminis-
guanethidine, which are discussed in this sec- tration. The major metabolites of reserpine are
tion. Other drugs which act by a mechanism sim- products of hydrolytic cleavage: methyl reser-
ilar to that of guanethidine and are marketed in pate and trimethoxybenzoic acid. Urinary excre-
some countries include: bethanidine, guanadrel, tion of reserpine metabolites accounts for 20 –
debrisoquin, guanacline, and guanochlor. They 40 % of the administered doses and fecal excre-
differ from guanethidine in potency, pharma- tion for less than 10 % [120], [121].
cokinetics, and some aspects of their pharmacol- The major side effects of reserpine are seda-
ogy. The mechanism of their antihypertensive tion, nasal stuffiness, and a depressive reaction.
action and the profile of their side effects are, Reserpine is also known to increase prolactin re-
however, not essentially different from those of lease. Adrenocorticotrophic hormone (ACTH)
guanethidine. Orthostatic hypotension is the ma- secretion is initially increased and subsequently
jor side effect which restricts their therapeutic abolished by reserpine. Gastric secretion is in-
usefulness. creased and formation of gastric ulcers is pro-
moted. The initial dose of reserpine utilized in
hypertension is 0.12 – 1.0 mg/d and the mainte-
nance dose is 0.25 mg or less. The use of reser-
pine alone has decreased because of the avail-
ability of drugs with less severe side effects. In
12 Antihypertensives

combination with thiazide diuretics reserpine is, in the region of parotid glands, and muscle weak-
however, still widely used. ness.

4.2.1.2. Guanethidine 4.2.2. α-Adrenoceptor Antagonists

α-Adrenoceptor antagonists, e.g., phentol-


amine, tolazoline, or yohimbine, were known for
many years [134], but were not used in the treat-
ment of hypertension, because of their question-
Guanethidine [55-65-2], [2-(hexahydro-1(- able effectiveness, rapid development of toler-
2H)-azocinyl)ethyl]guanidine, C10 H22 N4 , M r ance, and pronounced reflex tachycardia. The in-
198.31, mp 276 – 281 ◦ C. troduction of prazosin [135], [136] and demon-
Trade names: (sulfate salt) Ismelin, Guethi- stration of its usefulness in the treatment of hy-
ne, Iporal, Isobarin. pertension resulted in the development of vari-
The major pharmacological action of ous new α-adrenoceptor antagonists. The selec-
guanethidine is inhibition of release and of tive α1 -adrenoceptor antagonists are more ef-
uptake of norepinephrine by sympathetic fective in hypertension than antagonists affect-
nerve endings [122–125]. After treatment with ing both α1 - and α2 -adrenoceptors. The reduced
guanethidine, vasoconstrictor nerves do not effectiveness is most likely due to an inhibi-
provide reflex compensation to the erect po- tion of presynaptic α2 -adrenoceptor by nonse-
sition. Thus, the hypotension after guanethidine lective antagonists. This leads to an increase in
is largely orthostatically induced, which can neurotransmitter release and consequent tachy-
lead to undesirable effects, such as dizziness cardia and vasoconstriction, which oppose the
and syncope. In addition to its adrenergic neu- hypotensive effects of the drugs as a result of
ron blocking action, guanethidine has also a blockade of postsynaptic α1 -adrenoceptors. The
direct vasodilator activity, the mechanism of three drugs discussed here are relatively selec-
which may involve stimulation of vascular β- tive α1 adrenoceptor antagonists.
adrenoceptors [123], [126]. The hypotensive
action of guanethidine can be antagonized by
amphetamine, ephedrine, cocaine, or by tricyclic 4.2.2.1. Prazosin
antidepressants [123], [127].
By acute intravenous administrations to ani-
mals or humans, guanethidine produces a tran-
sient increase in arterial pressure which is fol-
lowed by hypotension. Peripheral vascular re-
sistance is not changed, while cardiac output
is decreased [128], [129]. In chronic oral ther- Prazosin [19216-56-9], 1-(4-amino-6,7-di-
apy guanethidine does not change cardiac output methoxy-2-quinazolinyl)-4-(2-furanylcarbon-
[130]. yl)piperazine, C19 H21 N5 O4 , M r 382.42, mp
Guanethidine is not completely absorbed 278 – 280 ◦ C, is water soluble.
from the gastrointestinal tract, with 20 – 25 % Trade names: Minipress, Hypovase, Sine-
of oral dose being recovered from the fe- tens.
ces [131]. 2-(6-Carboxyhexylamino)ethylguan- The blockade of α1 -adrenoceptors represents
idine and guanethidine N-oxide have been iden- the major mechanism of antihypertensive action
tified as metabolites of guanethidine [132], of prazosin [135–139]. The α1 -adrenoceptor an-
[133]. Guanethidine is given once a day at doses tagonist action of prazosin leads to arterial and
ranging from 12.5 to 150 up to 400 mg/d. The venous dilatation, a reduction in preload and af-
doses are adjusted by 10 or 25 mg at 2-week in- terload, and, consequently, to an improvement
tervals. In addition to orthostatic hypotension, in cardiac efficiency. Because of the indirect im-
side effects of guanethidine include water reten- provement in cardiac function, prazosin is also
tion, failure of ejaculation, diarrhea, tenderness
Antihypertensives 13

useful in the therapy of heart failure [140]. In α1 -adrenoceptors. It is a competitive antagonist


spite of pronounced vasodilatation, heart rate un- of norepinephrine and a noncompetitive antag-
dergoes no change or only a slight increase after onist of 5-HT and histamine. At doses higher
prazosin [138]. than required for lowering of arterial pressure
A consistent decrease in peripheral vascular (5 vs. 1 mg/kg, i.v. in cats) indoramin reduces
resistance differentiates prazosin from methyl- sympathetic nerve activity apparently by a cen-
dopa, clonidine or β-adrenoceptor antagonists. tral action [154–157]. Other pharmacological ef-
In spite of pronounced vasodilator action, pra- fects of indoramin include local anesthetic and
zosin in patients either decreases or has no effect antiarrhythmic effects [158], [159].
on plasma renin activity [141], [142]. The peak Indoramin is well absorbed orally. Its mean
plasma concentration of prazosin is reached half-life in humans was estimated to be 5.5 h
2 – 3 h after an oral dose and its half-life is 3.9 h [160]. The metabolism of indoramin is exten-
[143]. The major metabolites of prazosin in rats, sive. The major human metabolite was identified
dogs, and humans are 6-O- and 7-O-demethyl- as a conjugate of 6-hydroxyindoramin [161].
prazosin. Other minor metabolites have been Extensive clinical studies have been con-
identified [144], [145]. ducted with indoramin [162–164]. Indoramin
The antihypertensive activity of prazosin is is effective in the treatment of hypertension
comparable to, or better than, that of methyldopa at doses ranging from 20 to 150 mg/d. The
[146], hydralazine [147], or clonidine [148]. major side effect is sedation. Indoramin does
Prazosin is successfully used in combination not produce the “rebound” phenomenon after
with thiazide diuretics [149] or β-adrenoceptor discontinuation of therapy, nor syncope fol-
antagonists [150]. lowing the first dose. In addition to its anti-
The major side effect of prazosin is the so- hypertensive action, indoramin was reported to
called “first dose phenomenon”, a syncope in produce therapeutic benefits in headache and
patients receiving a 2-mg tablet as the first dose bronchial asthma.
[151]. The syncope can be avoided if the ini-
tial dose is reduced to 0.5 mg and given at bed-
time. The dose can then be increased gradually 4.2.2.3. Urapidil
over a 2-week period. Other reported side effects
include an increase in the frequency of urina-
tion and in the frequency of anginal attacks. The
usual dose of prazosin is 2 mg three times a day,
but the drug was used at total daily doses up to
20 mg.
Urapidil [34661-75-1], 6-({3-[4-(2-meth-
4.2.2.2. Indoramin oxyphenyl)-1-piperazinyl]propyl}amino)-
1,3-dimethyl-2,4(1H,3H)-pyrimidine-dione,
C20 H29 N5 O3 , M r 387.49, mp 156 – 158 ◦ C, is
poorly soluble in water, but readily soluble in
dilute acids.
Synthesis: [165].
Trade name: Ebrantil.
Urapidil lowers arterial pressure in hyperten-
Indoramin [26844-12-2], N-[1-[2-(1H- sive and normotensive animals. Its antihyper-
indol-3-yl)ethyl]-4-piperidinyl]benzamide, tensive activity is due to a combination of pe-
C22 H25 N3 O, M r 347.46, mp 208 – 210 ◦ C. The ripheral and central actions. The peripheral ac-
synthesis and pharmacological activity of indo- tion involves blockade of α1 -adrenoceptors in
ramin have been described [152–154]. vascular smooth muscle. By virtue of its central
Trade name: (hydrochloride salt) Baratol. action urapidil diminishes sympathetic outflow,
The mechanism of antihypertensive action of an effect which is probably mediated by central
indoramin involves primarily the blockade of
14 Antihypertensives

α-adrenoceptors and is essentially similar to that 4.2.3.1. Propranolol


of clonidine [166], [167].
Urapidil is well absorbed orally. The maxi-
mal blood levels are reached after 0.5 – 1 h after
oral administration of tablets and 2.5 – 4 h af-
ter administration of slow-release capsules. The
plasma half-life of urapidil in humans was esti-
mated as 2.5 – 3.7 h. Urapidil is metabolized by
Propranolol [525-66-6], 1-(isopropylamino)-
p-hydroxylation, O-demethylation of the meth-
3-(1-naphthyloxy)-2-propanol, C16 H21 NO2 ,
oxyphenyl ring, and N 1 -demethylation of the
M r 259.34, pK a 9.5, is moderately soluble in wa-
uracil residue. The same metabolites are found
ter and in ethanol. It is the most commonly used
in humans, dogs, rats, and mice. The p-hydroxyl-
β-adrenoceptor antagonist. The β-adrenoceptor
ation product has antihypertensive activity but is
blocking and the antihypertensive activities re-
less potent than urapidil [167], [168].
side in the (+)-isomer [172].
In animals, urapidil exhibits sedative activ-
Trade names: Inderal, Inderex, Docitor,
ity, while in humans, the major side effects in-
Tesnol, and many others.
clude dizziness, headache, and nausea. In the
Propranolol is not cardioselective; β 1 - as
treatment of hypertension, urapidil is effective
well as β 2 -adrenoceptors are blocked by the
at 30 – 90 mg/d given as slow-release capsules
drug. Propranolol has MSA but no ISA. β 2 -
[167], [169].
Adrenoceptor blocking activity can lead to
bronchoconstriction and propranolol is, there-
4.2.3. β-Adrenoceptor Antagonists fore, contraindicated in patients with bronchial
asthma. The MSA of propranolol is correlated
β-Adrenoceptor antagonists were introduced with local anesthetic as well as with the car-
in the treatment of hypertension on the basis diac depressant actions. At therapeutic doses the
of clinical observations that pronethalol, one MSA of propranolol is not prominent. The neg-
of the first β-adrenoceptor antagonists, low- ative dromotropic activity of propranolol is ev-
ered arterial pressure in patients with angina ident following acute as well as chronic admin-
pectoris [170]. The mechanism of antihyper- istration of the drug and is not opposed by the
tensive action of β-adrenoceptor blocking drugs ISA [173], [174].
is still controversial. Various hypotheses have Following acute intravenous administration
been critically reviewed [171]. Presently, re- to animals or humans, propranolol reduces car-
setting of baroreceptors and the inhibition of diac output, mean arterial pressure, heart work,
sympathetic transmission by blockade of pre- left ventricular dp/dt, and blood flow in most
synaptic β-adrenoceptors are considered the vascular beds. Total peripheral vascular resis-
most likely mechanisms of antihypertensive ac- tance is first increased but may be reduced during
tion, although lowering of plasma renin ac- chronic therapy. Plasma renin activity is consis-
tivity and a decrease in cardiac output by β- tently reduced by the drug.
adrenoceptor antagonists may play a contribu- Propranolol is nearly completely absorbed
tory role, particularly in the initial phase of ther- from the gastrointestinal tract but is a subject
apy. of a large first-pass effect [175]. Peak plasma
β-Adrenoceptor antagonists differ in po- levels of propranolol are seen within 1 – 3 h af-
tency, cardiac selectivity, intrinsic sympath- ter treatment [176]. Propranolol is extensively
omimetic activity (ISA), membrane stabilizing metabolized; 16 metabolites or conjugates have
action (MSA), duration of action, tissue distri- been identified. The major metabolites are naph-
bution, and metabolism. thoxylactic acid and 4-hydroxypropranolol (an
A large number of β-adrenoceptor antago- active metabolite) [177], [178].
nists were developed and marketed during the In the treatment of hypertension propranol-
last decade. The seven drugs discussed are more ol is usually used at 80 – 320 mg/d in four di-
commonly used and represent different sub- vided doses, although it was used at doses as
groups among β-adrenoceptor antagonists.
Antihypertensives 15

low as 40 mg and as high as 2 g/d. The drug and depression. Withdrawal reactions consist-
is commonly used in combination with a di- ing of headache, malaise, and palpitation were
uretic and/or a peripheral vasodilator drug [179], described for metoprolol. Thus, recommenda-
[180]. The side effects of propranolol include tions are to withdraw metoprolol slowly over a
bradycardia, A-V heart block, cardiac insuf- 10-d period. The initial dose of metoprolol in the
ficiency, bronchoconstriction, cold extremities, treatment of hypertension is 50 mg twice daily.
fatigue, vivid dreams, or even hallucinations. The maximal daily dose is 450 mg.
Rapid withdrawal of propranolol is not recom-
mended because it may lead to coronary insuf-
ficiency or myocardial infarction. 4.2.3.3. Nadolol

4.2.3.2. Metoprolol

Nadolol [42200-33-9], 5-[3-[(1,1-dimeth-


ylethyl)amino]-2-hydroxypropoxyl]-1,2,3,4-
tetrahydro-2,3-naphthalenediol, C17 H27 NO4 ,
M r 309.42, mp 124 – 136 ◦ C, pK a 9.67.
Metoprolol [37350-58-6], 1-[4-(2-methoxy- Trade names: Corgard, Solgol.
ethyl)phenoxy]-3-[(1-methylethyl)amino]-2- Like propranolol, nadolol is not cardioselec-
propanol, C15 H25 NO3 , M r 267.38. tive and has no ISA. Unlike propranolol, nadolol
Trade names: Beloc, Betaloc, Lopressor, Lo- has no MSA and, therefore, no direct cardiac de-
presor. pressant activity. The therapeutic advantages of
Unlike propranolol, metoprolol is relatively nadolol include ability to increase renal blood
specific for β 1 -adrenoceptors and is, therefore, flow, and a long duration of action (up to 24 h)
relatively free from bronchoconstrictor action [188–190]. The oral absorption of nadolol in hu-
[181], [182]. At therapeutic doses, metoprolol mans ranges from 20 to 34 %. Serum half-life
has no MSA, and therefore, has no local anes- of nadolol in humans is significantly longer than
thetic or direct myocardial depressant effects. that of other β-adrenoceptor antagonists and was
As a β 1 -adrenoceptor antagonist, metoprolol is estimated to equal 24 h [191].
equipotent to propranolol [183]. Metoprolol is In the treatment of hypertension nadolol is
well absorbed by oral administration but has given once a day at 40 – 240 mg [192]. In ad-
a substantial first-pass effect. Its plasma half- dition to its antihypertensive effect nadolol was
life is 3 – 4 h. Metoprolol is metabolized by O- shown to be effective in the treatment of angina
demethylation, subsequent oxidation, oxidative pectoris [193] and of certain types of ventricu-
deamination, and aliphatic hydroxylation. The lar arrhythmias [194]. Most of the side effects of
two metabolites, O-demethylated product and nadolol are similar to those of propranolol.
α-OH-metoprolol, have β 1 -adrenoceptor antag-
onist activity but are 5 times less potent than
4.2.3.4. Pindolol
metoprolol [184], [185].
In clinical studies, metoprolol was shown to
be equally effective as hydrochlorothiazide as an
antihypertensive agent [186] and either equiva-
lent or superior to other β-adrenoceptor antago-
nists [183]. In a double-blind study metoprolol
was shown to reduce the mortality in patients
with myocardial infarction [187]. Pindolol [13523-86-9], 1-(1H-indol-4-yl-
The side effects of metoprolol include tired- oxy)-3-[(1-methylethyl)amino]-2-propanol,
ness, headache, insomnia, nausea, heartburn, C14 H20 N2 O2 , M r 248.32, mp 171 – 173 ◦ C.
16 Antihypertensives

Like propranolol, pindolol is not cardioselec- anesthetic action. As a β-adrenoceptor antago-


tive, but unlike propranolol, pindolol has strong nist timolol is ca. 8 times more potent than prop-
ISA, which is responsible for its acute vasodila- ranolol. As a negative inotropic agent timolol is
tor effect and for absence of bradycardia. The 40 times less potent than propranolol [203–205].
MSA of pindolol is considerably weaker than Timolol is rapidly absorbed by oral admin-
that of propranolol. In spite of the absence of istration with 72 % of the dose excreted within
cardioselectivity, pindolol is less likely to impair 84 h. The plasma half-life of timolol in humans
pulmonary function than propranolol, probably was estimated as 5.5 h [206]. Five metabolites of
because of its strong ISA [195], [196]. timolol were identified in humans [205]. They
Pindolol is well absorbed by oral admin- have little or no β-adrenoceptor blocking activ-
istration and in comparison with other β- ity.
adrenoceptor antagonists, has a lower first-pass The clinical study in essential hypertension
effect (13 %) [197]. The plasma half-life of pin- indicated that timolol at 30 mg and propranol-
dolol is 3 – 4 h and the maximal plasma levels ol at 120 mg had comparable antihypertensive
are reached 1.5 – 2 h after oral administration to activity [207]. Timolol is used in the treatment
humans [198]. It has a more rapid onset of ac- of hypertension at 10 – 30 mg/d. In patients sur-
tion than propranolol and after withdrawal has viving after acute myocardial infarction, timolol
a longer carryover effect [199–201]. The dose was shown to reduce mortality and reinfarction
range of pindolol in the treatment of hyperten- rates [208]. The side effects of timolol include
sion is 10 – 30 mg/d. The drug was initially ad- tiredness, dizziness, increase in body mass, and
ministered t.i.d. but subsequently the majority an occasional increase in serum urea and creati-
of patients were found to respond equally well nine.
to once-a-day therapy [202].
The side effects of pindolol include in-
somnia and vivid dreams. Heart failure, heart 4.2.3.6. Acebutolol
block, bronchoconstriction, and circulatory dis-
turbances are rare with pindolol.

4.2.3.5. Timolol
Acebutolol [37517-30-9], N-(3-acetyl-4-{2-
hydroxy-3-[(1-methylethyl)amino]propoxy}-
phenyl)butanamide, C18 H28 N2 O4 , M r 336.43,
mp 128 – 129 ◦ C. Its synthesis and pharma-
cological activity were first described by
Wooldridge [209].
Timolol [26839-75-8], 1-[(1,1-dimethyl- Trade names: Sectral, Neptall, Prent.
ethyl)amino]-3-{[4-morpholinyl-1,2,5-thiadi- Unlike propranolol, acebutolol is cardiose-
azol-3-yl]oxy}-2-propanol, C13 H24 N4 O3 S, M r lective and has weak ISA and MSA [210–214].
316.42, is supplied as the hydrogen maleate salt, The major metabolite of acebutolol is diace-
C17 H28 N4 O7 S, M r 432.5, mp 201.5 – 202.5 ◦ C, tolol, RS-1-(2-acetyl-4-acetyl-amidophenoxy)-
which is soluble in water and ethanol and is 2-hydroxy-3-isopropylaminopropane), which
less lipophilic than propranolol. Unlike other has β-adrenoceptor blocking actions similar to
β-adrenoceptor antagonists, timolol is available those of acebutolol, but has no MSA [215]. The
as the (−)-isomer and not as a racemic mixture. half-life of acebutolol and its metabolite is ca.
Trade names: (hydrogen maleate salt) MK- 4 h. Acebutolol is metabolized in humans al-
950, Betim, Blocadren, Temserin, Timacor, most exclusively to diacetolol, 40 % of which is
Timolate, Timoptic, Timoptol. excreted unchanged [213].
Like propranolol, timolol is not cardioselec- The antihypertensive effect of acebutolol is
tive and has no ISA. At therapeutic doses timolol more pronounced in severe than in mild hy-
has little or no MSA and has practically no local pertension [216] and was confirmed in numer-
Antihypertensives 17

ous controlled trials [217], [218]. In hyperten- turbances, postural hypotension, impotence, and
sion acebutolol is given once or twice daily at scalp tingling.
doses ranging from 200 to 1200 mg/d. Side ef-
fects of acebutolol include blurred vision, bron-
chospasm, and bradycardia. 5. Smooth Muscle Relaxants
5.1. Ca2+ Channel Antagonists
4.2.3.7. Labetalol
The Ca2+ channel antagonists are a group
of vasodilators used extensively in the treat-
ment of angina pectoris (→ Calcium Antago-
nists, p. 519 ff.). However, their vasodilatory ac-
tions extend also to vascular beds other than the
coronary and, consequently, their usefulness in
Labetalol [36894-69-6], 2-hydroxy-5-{1- hypertension has become apparent. The group
hydroxy-2-[(1-methyl-3-phenylpropyl)amino]- classification emanates from the fact that these
ethyl}benzamide, C19 H24 N2 O3 , M r 328.41; the agents potently inhibit the entry of Ca2+ into
hydrochloride salt, C19 H25 ClN2 O3 , is soluble vascular smooth muscle [227–229], although
in water and ethanol. Labetalol has two asym- other mechanisms may contribute to their ef-
metric centers and exists as a mixture of four fectiveness as well [230], [231]. Considerable
isomers in equal proportions. chemical diversity is found in the representatives
Trade names: (hydrochloride salt) Presdate, of the Ca2+ channel antagonists [232], [233].
Trandate. Nifedipine, nitrendipine, verapamil, and dilti-
Labetalol differs from other β-adrenoceptor azem have been studied most extensively in the
antagonists in its ability to block not only β- treatment of hypertension; therefore, the empha-
but also α-adrenoceptors, although it is more sis here is placed on these agents.
potent in blocking β-adrenoceptors. As a β-
adrenoceptor antagonist, labetalol is not car-
dioselective, has weak ISA, but has clearly de- 5.1.1. Nifedipine
tectable MSA. As an α-adrenoceptor antago-
nist, labetalol is selective for α1 -adrenoceptors.
Because of its α1 -adrenoceptor blocking activ-
ity labetalol decreases peripheral vascular re-
sistance, particularly at higher doses. The anti-
hypertensive activity of labetalol is likely to be
due to α- as well as β-adrenoceptor blocking
effects [219–221]. Nifedipine [21829-25-4], 1,4-dihydro-2,6-
Following oral administration labetalol is dimethyl-4-(2-nitrophenyl)-3,5-pyridinecarb-
well absorbed and excreted in the form of glu- oxylic acid dimethyl ester, C17 H18 N2 O6 , M r
curonide conjugates. The major metabolite is 346.34, mp 172 – 174 ◦ C, is a yellow crystalline
the alcoholic glucuronide of labetalol [221]. compound, practically insoluble in water, but
Plasma labetalol levels are well correlated with soluble in organic solvents, such as ethanol,
its pharmacological actions [222]. Labetalol ex- polyethylene glycol, and chloroform. In solu-
periences high first-pass metabolism [223]. tion, it decomposes quite rapidly under normal
Extensive clinical studies with labetalol were laboratory light.
published [224–226]. In humans labetalol low- Trade names: BAY a 1040, Adalat, Procar-
ers arterial pressure without any significant ef- dia, Duranifin, Pidilat.
fect on resting heart rate or on cardiac output. It The structure – activity requirements are re-
is used in hypertension at 200 – 2000 mg/d, usu- ported for several muscle systems, including
ally in two divided doses. Side effects of labetalol vascular, cardiac, and nonvascular smooth mus-
include tiredness, dizziness, gastrointestinal dis- cle (see reviews [230], [234] ). Specific require-
ments are the following: (1) the 1,4-dihydro-
18 Antihypertensives

pyridine (DHP) ring is essential; (2) an unsub- Metabolism occurs primarily in the liver, with
stituted NH-group on the DHP-ring; (3) 2,6- the majority being excreted in the urine and fe-
substituents on the DHP-ring should be lower ces as polar metabolites [260], [263].
alkyl substituents, but an NH2 group is accept- The adverse reactions during therapy include
able; (4) ester groups in the 3- and 5-positions of headache, flushing, and nausea. In some pa-
the DHP-ring can be substituted to change the tients, a localized ankle edema has been ob-
vascular selectivity and duration of action; (5) served, which may be attributed to a change
ester groups longer than COOMe will increase in capillary permeability or perfusion pressure
activity; (6) the aryl substituent (i.e., phenyl [264]. The oral dosage form is presently ap-
ring) is optimal in the 4-position of the DHP- proved for use in angina pectoris.
ring; (7) position of the substituent on the phenyl
ring is critical, i.e., para results in decreased ac-
tivity, while ortho or meta result in increased ac- 5.1.2. Nitrendipine
tivity; (8) stereoselectivity is obtained by differ-
ent ester substitutions on C-3 and C-5 of DHP-
ring [235–237].
Nifedipine lowers blood pressure in many ex-
perimental models of hypertension [238], [239].
It is efficacious in mild to moderate essentially
hypertensive patients [240], [241] and in patients
exhibiting severe hypertension [242], [243]. Fol-
lowing oral administration, the antihypertensive Nitrendipine [39562-70-4], 1,4-dihydro-2,6-
responses are rapid in onset (30 – 60 min), with a dimethyl-4-(3-nitrophenyl)-3,5-pyridinedicarb-
duration of about 3 h. Nifedipine exhibits dimin- oxylic acid ethyl methyl ester], M r 360.4, forms
ished activity in normotensive subjects in com- yellow crystals with solubility properties simi-
parison to hypertensives [240], [244–247]. lar to nifedipine; however, nitrendipine is less
The antihypertensive effect of nifedipine is light sensitive, with a decomposition half-life of
due to arterial vasodilatation and a reduced to- about 7 h in laboratory light. It exhibits Ca2+
tal peripheral resistance [240], [244], [248–252]. channel antagonism and produces antihyper-
Compensatory increases in heart rate, plasma tensive effects in a number of animal models.
norepinephrine, and renin levels are found after It is more potent than nifedipine on vascular
single doses of nifedipine; however, these eleva- strips in vitro, and has a longer duration of anti-
tions usually subside during long-term therapy hypertensive action in the renal hypertensive
[253–256]. dog model [265–275].
Combined therapy of nifedipine with the β- Trade names: Bayotensin, Baypress.
adrenoceptor antagonists, propranolol or meto- Synthesis: [268].
prolol, slightly enhanced the antihypertensive In clinical studies, single or repeated daily
response in humans [240], [255]. The addition of oral doses of nitrendipine (5 – 40 mg) effec-
methyldopa to nifedipine in severely hyperten- tively lower arterial pressure. The onset of ac-
sive patients increased the antihypertensive re- tion and peak effects are obtained at 15 min and
sponse and reduced blood pressure fluctuations at 60 – 90 min, respectively. The duration of ac-
between dosing [242]. tion is approximately 8 h following a single oral
Sodium and water retention are not a prob- dose, although, with long-term therapy, blood
lem during nifedipine treatment. Urinary vol- pressure can be controlled for up to 24 h with
ume and sodium excretion are elevated in hy- one dose [276–279]. In hemodynamic studies,
pertensive patients during nifedipine treatment nitrendipine lowered total peripheral resistance,
[257], [258]. Should volume retention occur it but did not significantly affect cardiac output
can be controlled by diuretic therapy [241]. or heart rate [279], [280]. Plasma renin activ-
Nifedipine is bound extensively to plasma ity and catecholamines levels are increased by
proteins (> 90 %) [259], [260]. The plasma nitrendipine, but aldosterone levels are not sig-
concentration of nifedipine correlates well nificantly elevated [280].
to the hemodynamic responses [261], [262].
Antihypertensives 19

In rats and humans, the metabolism of ni- 296]. Constipation is the most prevalent side ef-
trendipine involves dehydrogenation, deesterifi- fect of verapamil therapy [283].
cation, and hydroxylation reactions. The identi-
fied metabolites exhibit essentially no antihyper-
tensive activity [265]. Adverse reactions in hu- 5.1.4. Diltiazem
mans include headache, flushing, and nausea.

5.1.3. Verapamil

Diltiazem [42399-41-7], 3-(acetyloxy)-5-


[2-(dimethylamino)ethyl]-2,3-dihydro-2-(4-
methoxyphenyl)-1,5-benzothiazepin-4(5H)-
Verapamil [52-53-9], α-(3-{[2-(3,4-dimeth- one, C22 H26 N2 O4 S, M r 414.52, is a white crys-
oxyphenyl)ethyl]-methylamino}propyl)-3,4- talline powder, freely soluble in water, methanol,
dimethoxy-α-(1-methylethyl)benzeneaceto- or chloroform. The chemical synthesis of dilti-
nitrile, C27 H38 N2 O4 , M r 454.59, is a white azem and related substances has been reported
crystalline powder, soluble in water, chloro- [297], [298].
form, and methanol; the compound is not light Trade names: Cardizem Herbesser, Dilzem.
sensitive. It is a Ca2+ channel antagonist which The vasodilator structure – activity relation-
is unrelated chemically to nifedipine and ni- ships have been reported and summarized [299–
trendipine. 301]. Diltiazem is the d-cis isomer. The trans
Trade names: Isoptin, Calan, Cardibeltin, isomeric forms exhibit much lower vasodila-
Cordilox, Vasolan. tor activity. Other substitutions which result
Comprehensive reviews and studies on the in lower activity include replacement of the
pharmacology of verapamil have been published methoxy group with a hydroxyl group or addi-
[281–287]. tional alkoxy substituents on the aromatic ring.
In clinical studies, verapamil lowered blood In addition, removal of the alkyl-aminoalkyl
pressure in patients with essential hypertension group at N5 will significantly reduce the activ-
[288–290]. The dosage ranged between 120 to ity, as will dealkylation or quaternization of the
160 mg, given three times daily. Total peripheral terminal nitrogen. Other alterations, such as re-
resistance was decreased and cardiac output was placement of the methoxy group by a p-methyl
elevated by verapamil. Heart rate was either un- and chlorine substitution at the 7-position of a
changed or decreased during therapy. The anti- condensed aromatic ring, do not reduce activity.
hypertensive efficacy was directly related to pre- The antihypertensive activity of diltiazem has
treatment levels of blood pressure and inversely been demonstrated in DOCA, renal, and sponta-
related to plasma renin levels [290]. The va- neously hypertensive rats [302–304]. Diltiazem
sodilator potency when given directly into the increased cardiac output, significantly elevated
forearm circulation was greater in hypertensive cardiac and skeletal muscle blood flow, and pro-
patients in comparison with normotensive sub- duced lesser flow increases to the mesenteric,
jects. In addition, the increase in forearm blood cerebral, and renal vascular beds. Flow to the
flow correlated positively with basal concentra- skin is usually unchanged or reduced by dilti-
tions of plasma epinephrine but not with plasma azem [301], [305–307].
levels of renin or aldosterone [291]. Verapamil In humans, diltiazem is effective in mild-to
fails to elevate plasma catecholamine, renin, or moderate hypertension at oral doses ranging bet-
aldosterone levels. The mechanism responsible ween 180 – 270 mg/d. The reduction in blood
for this effect is unknown; however, possibil- pressure (−10 to −15 %) was accompanied by
ities might involve catecholamine depletion, a an increase in cardiac output. Similar to preclin-
decreased neuronal norepinephrine release, or ical studies, heart rate was not increased and
depressed baroreceptor reflex sensitivity [292– plasma catecholamines were only moderately
20 Antihypertensives

increased [308]. In another study, intravenous Hydralazine [86-54-4], 1-(2H)-phthalazinone


diltiazem (0.2 mg/kg plus 0.1 mg/kg-min, i.v.) hydrazone, C8 H8 N4 , M r 160.18, mp 172 –
dilated both arterioles and larger conduit arter- 173 ◦ C, is a yellow crystalline substance, soluble
ies in hypertensive patients. These results con- in acetic acid. Hydralazine has been available for
trasted with dihydralazine which dilated arteri- the treatment of hypertension for about 30 years.
oles, but constricted larger arteries [309]. Trade names: Apresoline HCl, Hydralazine
Details on the mechanism of action and other HCl; (combination with hydrochlorothiazide)
pharmacological properties of diltiazem have Apresoline-Esidrix, Apresozide, Hydrap-ES;
been summarized [305]. Metabolism occurs pri- (with reserpine) Serpasil-Apresoline; (with hy-
marily in the liver. Primary biotransformation drochlorothiazide plus reserpine) Ser-Ap-Es.
entails demethylation of the terminal nitrogen, Hydralazine lowers arterial pressure in many
cleavage of the p-methoxy group, and oxida- experimental models of hypertension. Certain
tive hydroxylation of the aromatic rings. The 3- structure requirements are necessary for maxi-
hydroxy compound is a major metabolite which mal activity. Among the more important require-
exhibits some pharmacological activity [301], ments are the presence of a free NH2 group and
[310]. the placement of the hydrazino-group in the 1-
position of the phthalazine moiety for optimal
duration of action. The activity is affected mini-
5.2. Vasodilators with Other mally with the substitution of a carbon in the ph-
Mechanisms of Action thalazine or a pyridine replacement of the benz-
ene ring [311].
Vasodilator drugs with a direct action on vas- Antihypertensive actions of hydralazine re-
cular smooth muscle were used in the therapy sult primarily from arteriolar relaxation and a
of hypertension long before the discovery of decreased peripheral resistance, with essentially
Ca2+ channel antagonists. Among vasodilator no effects on venous tone. Heart rate and car-
drugs hydralazine is the most commonly used diac output are increased by hydralazine. The
drug, although its precise mechanism of action cardiotonic action results primarily from re-
is still controversial. The common pharmacolog- flex adjustments to the decreased arterial pres-
ical properties of these drugs can be summarized sure (baroreceptor reflex) [312] or quite possibly
as follows: from elevation in central venous pressure (Bain-
bridge reflex) [313]. In addition, a direct car-
1) Their hypotensive activity is independent of diac stimulant effect has been observed in anes-
the sympathetic tone. thetized dogs by some [314], but not by other
2) They antagonize the vasoconstrictor effects investigators [313]. The direct and reflex actions
of various endogenous substances includ- of hydralazine are antagonized by propranolol.
ing norepinephrine, serotonin, angiotensin Cardiac hypertrophy occurring in SHR is not at-
II, and K+ . tenuated by hydralazine, whether given alone or
3) Their site of action is at the membrane of in combination with propranolol [315]. Exten-
vascular smooth muscle, at the contractile sive pharmacological studies with hydralazine
elements, or on the enzymatic processes in- were published [316–328].
volved in the delivery of energy for the con- Clinically, the antihypertensive effect of
tractile activity. hydralazine is enhanced by the addition of
4) Their common side effects include tachy- β-adrenoceptor antagonists, reserpine, guan-
cardia, salt and water retention, myocardial ethidine, methyldopa, clonidine, or diuretics
necrosis, and hyperglycemia. [329], [330]. When given in combination with
venodilators (e.g., isosorbide dinitrate or nitro-
glycerin) hydralazine has proved especially use-
5.2.1. Hydralazine ful in the treatment of congestive heart failure
[331].
Antihypertensives 21

5.2.2. Diazoxide Minoxidil [38304-91-5], 6-(1-piperidinyl)-2,4-


pyrimidinediamine-3-oxide, C9 H15 N5 O, M r
209.25, mp 248 ◦ C, 259 – 261 ◦ C (decomp.), is a
white crystalline solid. Minoxidil is primarily a
dilator of arterial vasculature, thereby lowering
total vascular resistance and blood pressure.
Trade name: Loniten.
Diazoxide [364-98-7], 7-chloro-3-meth- Extensive literature on pharmacology of mi-
yl-2H-1,2,4-benzothiadicazine-1,1-dioxide, noxidil exists [344–353].
C8 H7 ClN2 O2 S, M r 230.70, mp 330 – 331 ◦ C, is Minoxidil is approved primarily for use in
chemically related to chlorothiazide and other severely hypertensive patients who do not re-
benzothiadiazine diuretics, but it exhibits no spond to maximal therapy with a diuretic or other
diuretic activity. It is an effective antihyper- antihypertensive agents, or in patients where
tensive agent in spontaneous, DOCA, and re- side effects preclude other drug therapy. It is
nal hypertensive rat models [332], [333]. Anti- also useful in moderately hypertensive patients
hypertensive doses of diazoxide, when given [354], those with impaired renal function [355],
intravenously, range between 2 and 10 mg/kg in but is used rarely in hypertensive emergencies
animal studies. The duration of action is dose- because of its availability in oral dosage form
related and ranges between 0.5 – 4 h. only and its long onset of action [345]. Minoxi-
Trade name: Hyperstat I.V. dil is combined with β-adrenoceptor blockade to
When given intravenously, diazoxide reduces reduce effects of reflex sympathetic activation. It
total peripheral resistance and increases stroke is also effective when combined with other cen-
volume and cardiac output [334]. trally acting sympatholytics, such as clonidine
In humans, heart rate and force of left ven- [356], or methyldopa, α-adrenoceptor antago-
tricular contractions are increased by diazoxide, nists, or neuronal blocking agents [357]. Capto-
while left ventricular systolic and end-diastolic pril has been used as an adjunct to minoxidil in
pressures are decreased. β-Adrenoreceptor refractory hypertensive cases [358]. Because of
blockade attenuates the elevations in heart rate sodium and water retention, a diuretic is given in
and in left ventricular dp/dt, but augments the almost all cases; however, this may not be nec-
reduction in left ventricular systolic pressure essary in patients with less severe hypertension
[335]. The mechanism of action of diazoxide [345].
remains to be fully elucidated [336–340]. The use of minoxidil is limited to a rela-
Hyperglycemia is observed during diazoxide tively small population of hypertensive patients
therapy. This effect is attenuated by propranol- because of the incidence of hypertrichosis, and
ol [341] and the ganglionic blocker chlorison- potential for the occurrence of atrophic and is-
damine [342]. The propensity of diazoxide to chemic lesions of the heart [359]. The hyper-
cause hyperglycemia, and Na+ and fluid re- trichosis occurs in about 80 % of the patients
tention has restricted its clinical use to acute treated. It occurs about 3 – 6 weeks after the on-
life-threatening hypertensive emergencies. The set of therapy and requires about 1 – 6 months
dosage is titrated on the basis of individual for recovery after discontinuation of minoxidil.
responsiveness, but ranges between 100 and Cell culture experiments showed that minoxidil
300 mg, i.v. [343]. may produce the hypertrichosis by a direct ac-
tion on epidermal cells [360].

5.2.3. Minoxidil
5.2.4. Sodium Nitroprusside

Sodium nitroprusside [14402-89-2] is used


as the dihydrate, Na2 Fe(CN)5 NO · 2 H2 O, M r
216.91, readily soluble in water, but decomposes
in light.
22 Antihypertensives

Trade names: Nipride, Nitropress. Ca2+ channels are likely to be developed dur-
Nitroprusside is not absorbed orally and thus ing the next decade. Drugs interfering with in-
must be given by systemic infusion. Vasodila- tracellular Ca2+ translocation, e.g., calmodulin
tory effects are exerted on both the arterial and inhibitors, also can be conceivably useful in the
venous sides of the vasculature. Infusion of treatment of hypertension.
sodium nitroprusside at 1 to 100 µg kg−1 min−1 The central control of arterial pressure by
produces a uniform fall in systolic and diastolic drugs is receiving renewed attention. The recog-
blood pressure. The onset of action is usually nition of the importance of various neurotrans-
rapid (< 60 s) and the effect is well-maintained mitters, e.g., 5-HT, GABA, dopamine, in the
during the infusion. Recovery of blood pres- central regulation of cardiovascular functions is
sure occurs rapidly following cessation of drug likely to lead to novel antihypertensive drugs
[361]. This most likely is the result of rapid cel- with central mechanism of action. Modification
lular degradation of sodium nitroprusside. Be- of the synthesis or release of various neuropep-
cause of this rapid recovery the magnitude of tides may also represent a future mechanism of
the hypotensive response can be controlled eas- antihypertensive action. The role of vasopressin
ily by varying the infusion rate. Tolerance to in the cerebral as well as peripheral control of
repeated sodium nitroprusside infusions occurs vascular tone has received renewed attention,
rarely [362]. Other pharmacological effects have and vasopressin antagonists can conceivably be
been described [363–367]. useful in the treatment of hypertension.
The vasodilator activity of sodium nitroprus- The importance of the “natriuretic hormone”
side is attributed to the NO-group (nitroso) [368– in the control of Na+ levels in vascular smooth
370]. The vasorelaxant effects do not directly in- muscle cell and consequently of Ca2+ levels is
volve α- or β-adrenoceptors and are quite fully now being recognized, and drugs controlling the
manifested when vessels are contracted by an- release or the effects of “natriuretic hormone”
giotensin or vasopressin. Sodium nitroprusside may be useful in the treatment of hypertension.
may reduce the transsarcolemmal influx of cal-
cium or enhance calcium efflux by stimulating
Na+ −K+ ATPase, although quite disparate data
have been found in both regards [371–373]. El- 7. Summary and Conclusions
evations in cellular levels of cyclic GMP are in-
timately involved in the vasodilatory responses In spite of the discovery of two new classes
[369], [370]. of antihypertensive drugs, inhibitors of the
A final metabolic product of sodium nitro- angiotensin converting enzyme and Ca2+
prusside is thiocyanate. Toxicity during pro- channel antagonists, the well-established anti-
longed infusions is usually due to thiocyanate hypertensive drugs, such as diuretics and β-
accumulation (i.e., hypoxia, nausea, tinnitus) adrenoceptor antagonists, are still being widely
and necessitates discontinuation of treatment. used.
Diuretics which do not produce hypokalemia,
hyperuricemia, hyperglycemia, or hyperlipopro-
6. Perspectives in the Development of teinemia, or diuretics with an additional direct
vasodilator action are still in demand and may
Antihypertensive Drugs
be introduced in the near future.
A large number of new antihypertensive drugs The novelty of the mechanism of action as
have been introduced during the last 30 years. well as the clinical efficacy and safety will help
These drugs were usually justified by an im- to expand the market for converting enzyme in-
proved side effect profile and/or by a more phys- hibitors. Their apparent usefulness in the treat-
iological mechanism of action. This trend is ment of heart failure will also contribute to the
likely to continue during the next decade. Many expansion of their market share.
new inhibitors of angiotensin converting enzyme Among drugs which interfere with the sym-
have been introduced. The effectiveness of Ca2+ pathetic nervous system, α1 -, β-, and α1 /β-
channel antagonists in the treatment of hyper- adrenoceptor antagonists are widely used in the
tension was recognized and novel inhibitors of treatment of hypertension. The advantages of
Antihypertensives 23

α1 -adrenoceptor antagonists (e.g., prazosin) in- 15. W. E. Barrett, J. J. Chart, A. A. Renzi, Arch. Int.
clude increase in blood flow to vital organs Pharmacodyn. Ther. 131 (1961) 325 – 338.
and lowering of peripheral vascular resistance. 16. R. V. Ford, Curr. Ther. Res. 2 (1960)
βAdrenoceptor antagonists control arterial pres- 422 – 429.
sure with minimal side effects, and reduce 17. A. Scriabine, B. Korol, B. Kondratas, M. Yu et
mortality in patients with myocardial infarc- al., Proc. Soc. Exp. Biol. Med. 107 (1961)
tion. It remains to be seen whether new α1 /β- 864 – 872.
adrenoceptor antagonists (e.g., labetalol) will 18. H. Horstmann, H. Wollweber, K. Meng,
combine the benefits of both types of drugs. Arzneim.-Forsch. 17 (1967) 653 – 659.
19. K. Meng, G. Kroneberg, Arzneim.-Forsch. 17
With the discovery of the antihypertensive ac-
(1967) 659 – 671.
tion of Ca2+ channel antagonists the interest in
20. R. Rutsaert, M. Fernandes in A. Scriabine
the direct acting smooth muscle relaxants has (ed.): New Drugs Annual: Cardiovascular
been renewed and drugs which interfere with Drugs, vol. 1, Raven Press, New York 1983,
vascular smooth muscle tone and affect intra- pp. 49 – 67.
cellular calcium translocation are likely to be 21. E. G. Stenger, H. Wirz, R. Pulver, Schweiz.
introduced in the near future. Med. Wochenschr. 89 (1959) 1126 – 1130.
22. C. M. Kagawa, F. M. Sturtevant, C. G. Van
Arman, J. Pharmacol. Exp. Ther. 126 (1959)
8. References 123 – 130.
23. L. S. Watson, G. M. Fanelli, H. F. Russo, C. S.
1. E. J. Cragoe (ed.): Diuretics: Chemistry, Sweet et al. in A. Scriabine, C. S. Sweet (ed.):
Pharmacology and Medicine, J. Wiley & New Antihypertensive Drugs, Spectrum
Sons, New York 1983. Publications, Inc., New York 1976,
2. T. H. Maren, Trans. N.Y. Acad. Sci. 15 (1952) pp. 307 – 321.
53. 24. G. M. Fanelli, Jr., D. L. Bohn, A. Scriabine,
3. F. C. Novello, J. M. Sprague, J. Am. Chem. K. H. Beyer, J. Pharmacol. Exp. Ther. 200
Soc. 79 (1957) 2028 – 2029. (1977) 402 – 412.
4. K. H. Beyer, Ann. N.Y. Acad. Sci. 71 (1958) 25. W. B. Abrams, J. D. Irvin, J. A. Tobert, R. K.
363 – 379. Ferguson et al. in A. Scriabine (ed.): New
5. E. D. Fries, A. Wanko, I. M. Wilson, A. E. Cardiovascular Drugs, Raven Press, New
Parrish, JAMA 166 (1958) 137 – 140. York, 1985, pp. 1 – 20.
6. J. E. Baer, C. B. Jones, S. A. Spitzer, H. F. 26. E. Moller, H. Horstmann, K. Meng, D. Loew,
Russo, J. Pharmacol. Exp. Ther. 157 (1967) Experientia 33 (1977) 382 – 383.
472 – 485. 27. B. Garthoff, M. Hanisch in A. Scriabine (ed.):
7. V. D. Wiebelhaus, F. T. Brennan, G. Sosnowski New Drugs Annual: Cardiovascular Drugs,
et al., Arch. Int. Pharmacodyn. Ther. 169 vol. 2, Raven Press, New York 1984,
(1967) 429 – 457. pp. 51 – 70.
8. A. C. Guyton, R. D. Manning, J. E. Hall, R. A. 28. C. S. Sweet, E. H. Blaine in M. J. Antonaccio
Norman et al., J. Cardiovasc. Pharmacol. 6 (ed.): Cardiovascular Pharmacology, 2nd ed.,
(1984) S 151 – S 161. Raven Press, New York 1984, pp. 119 – 154.
9. M. P. Blaustein, Am. J. Physiol. 232 (1977) 29. M. A. Ondetti, B. Rubin, D. W. Cushman,
C 165 – C 173. Science 196 (1977) 441 – 444.
10. L. Poston, H. H. Gray, A. Crowther, H. C.
30. D. W. Cushman, M. A. Ondetti, Prog. Med.
Dittrich et al., J. Cardiovasc. Pharmacol. 6
Chem. 17 (1980) 41 – 104.
(1984) S 16 – S 20.
31. B. Rubin, R. J. Laffan, D. G. Kotler, E. H.
11. M. B. Strauss, H. Southworth, Bull. John
O’Keefe et al., J. Pharmacol. Exp. Ther. 204
Hopkins Hosp. 63 (1938) 41 – 45.
(1978) 271 – 280.
12. R. Muschaweck, P. Hajdú, Arzneim.-Forsch.
32. R. J. Laffan, M. E. Goldberg, J. P. High, T. R.
14 (1964) 44 – 47.
Schaeffer et al., J. Pharmacol. Exp. Ther. 204
13. K. H. Beyer, J. E. Baer, J. K. Michaelson, H. F.
(1978) 281 – 288.
Russo, J. Pharmacol. Exp. Ther. 147 (1965)
33. R. J. Bengis, T. G. Coleman, D. B. Young,
1 – 22.
14. G. de Stevens, L. H. Weiner, A. Halamandaris, R. E. McCaa, Circ. Res. 43 (1978) Suppl. 1,
S. Ricca, Experientia 14 (1958) 463. I 45 – I 53.
24 Antihypertensives

34. C. S. Sweet, P. T. Arbegast, S. L. Gaul, E. H. 56. D. M. Gross, C. S. Sweet, E. H. Ulm, E. P.


Blaine et al., Eur. J. Pharmacol. 76 (1981) Backlund et al., J. Pharmacol. Exp. Ther. 216
167 – 176. (1981) 552 – 557.
35. M. L. Cohen, K. D. Kurz, J. Pharmacol. Exp. 57. C. S. Sweet, D. M. Gross, P. T. Arbegast, S. L.
Ther. 220 (1982) 63 – 69. Gaul et al., J. Pharmacol. Exp. Ther. 216
36. T. Unger, D. Ganten, R. E. Lang, Trends (1981) 558 – 566.
Pharmacol. Sci. 4 (1983) 514 – 519. 58. J. Biollaz, M. Burnier, G. A. Turini, B. B.
37. M. J. Antonaccio, L. Kerwin, Hypertension 3 Brunner et al., Clin. Pharmacol. Ther. 29
(1981) Suppl. 1, I 54 – I 62. (1981) 665 – 670.
38. Y. Imai, K. Abe, M. Seino, T. Haruyama et al., 59. R. K. Ferguson, P. H. Vlasses, J. D. Irvin, B. N.
Hypertension 4 (1982) 444 – 451. Swanson et al. J. Clin. Pharmacol. 22 (1982)
39. R. Hatton, D. P. Clough, J. Cardiovasc. 281 – 289.
Pharmacol. 4 (1982) 116 – 123. 60. H. Gavras, B. Waeber, I. Gavras, J. Biollaz et
40. A. DeJonge, B. Wilffert, H. O. Kalkman, al., Lancet 2 (1981) 543 – 547.
J. C. A. van Meel et al., Eur. J. Pharmacol. 74 61. H. Gavras, J. Biollaz, B. Waeber, H. R.
(1981) 385 – 386. Brunner et al., Clin. Exp. Hypertens. 4 (1), (2)
41. C. Richer, M. P. Doussau, J. F. Giudicelli, Eur. (1982) 303 – 314.
62. F. G. Dunn, W. Oigman, H. O. Ventura, F. H.
Heart J. 4 (1983) Suppl. G, 55 – 60.
Messerli et al., Am. J. Cardiol. 53 (1984)
42. T. Unger, I. Kaufmann-Bühler, B. Schölkens,
105 – 108.
D. Ganten, Eur. J. Pharmacol. 70 (1981)
63. P. W. deLeeuw, R. P. L. M. Hoogma, G. A. W.
467 – 478.
van Soest, P. T. Tchang et al., J. Cardiovasc.
43. R. E. McCaa, J. E. Hall, C. S. McCaa, Circ.
Pharmacol. 5 (1983) 731 – 736.
Res. 43 (1978) Suppl. 1, I 32 – I 39.
64. S. Morioka, G. Simon, J. N. Cohn, Clin.
44. V. S. Murthy, T. L. Waldron, M. E. Goldberg,
Pharmacol. Ther. 34 (1983) 583 – 589.
Circ. Res. 43 (1978) Suppl. 1, I 40 – I 45. 65. C. Richer, M. P. Doussau, J. F. Giudicelli, Eur.
45. K. Ito, H. Koike, M. Miyamoto, H. Ozaki et J. Pharmacol. 79 (1982) 23 – 29.
al., J. Pharmacol. Exp. Ther. 219 (1981) 66. D. M. Shoback, G. H. Williams, S. L. Swartz,
520 – 525. R. O. Davies et al., J. Cardiovasc. Pharmacol.
46. B. Rubin, M. J. Antonaccio in A. Scriabine 5 (1983) 1010 – 1018.
(ed.): Pharmacology of Antihypertensive 67. O. Kohlmann, M. Bresnahan, H. Gavras,
Drugs, Raven Press, New York 1980, Hypertension 6 (1984) Suppl. 1, I 1 – I 6.
pp. 21 – 42. 68. A. Scriabine, D. G. Taylor, E. Hong, Prog.
47. E. E. Muirhead, R. L. Prewitt, B. Brooks, W. L. Drug Res. 26 (1982) 353 – 371.
Brosius, Circ. Res. 43 (1978) Suppl. 1, 69. A. Scriabine, B. V. Clineschmidt, C. S. Sweet,
I 53 – I 59. Annu. Rev. Pharmacol. Toxicol. 16 (1976)
48. D. B. Case, S. A. Atlas, P. A. Sullivan, J. H. 113 – 123.
Laragh, Circulation 64 (1981) 765 – 771. 70. T. L. Sourkes, Arch. Biochem. Biophys. 51
49. W. B. Lee, M. J. Ismay, E. R. Lumbers, Circ. (1954) 444 – 456.
Res. 47 (1980) 286 – 292. 71. L. Gillespie, J. A. Oates, J. R. Crout, A.
50. W. H. Van Gilst, P. A. DeGraeff, J. H. Kingma, Sjoerdsma, Circulation 25 (1962) 281 – 291.
H. Wesseling et al., Eur. J. Pharmacol. 100 72. C. C. Porter, M. L. Torchiana, C. A. Stone in F.
(1984) 113 – 117. Gross (ed.): Antihypertensive Agents,
51. B. G. Zimmerman, P. C. Wong, G. K. Springer-Verlag, Berlin-Heidelberg-New York
Kounenis, E. J. Kraft, Am. J. Physiol. 243 1977, pp. 269 –297.
(1982) H 277 – H 283. 73. M. Henning, P. A. van Zwieten, J. Pharm.
52. S. Chiba, C. P. Quilley, J. Quilley, J. C. McGiff, Pharmacol. 20 (1968) 409 – 417.
Eur. J. Pharmacol. 83 (1982) 243 – 252. 74. A. Scriabine in A. Scriabine (ed.):
53. T. Taylor, T. J. Moore, N. K. Hollenberg, G. H. Pharmacology of Antihypertensive Drugs,
Williams, Hypertension 6 (1984) 92 – 99. Raven Press, New York 1980, pp. 43 – 54.
54. W. B. Abrams, R. O. Davies, R. K. Ferguson, 75. G. Onesti, A. N. Brest, P. Novack, H.
Fed. Proc. 43 (1984) 1314 – 1321. Kasparian et al., Am. Heart J. 67 (1964)
55. C. S. Sweet, E. H. Ulm in A. Scriabine (ed.): 32 – 38.
76. S. Mohammed, T. E. Gaffney, A. C. Yard, H.
New Drugs Annual: Cardiovascular Drugs,
Gomez, J. Pharmacol. Exp. Ther. 160 (1968)
vol. 2, Raven Press, New York 1984, pp. 1 – 17.
300 – 307.
Antihypertensives 25

77. A. Cohen, J. S. Maxmen, M. Ragheb, H. 98. G. Scholtysik, H. Lauener, E. Eichenberger, E.


Baleiron et al., J. Clin. Pharmacol. 7 (1967) Bürki et al., Arzneim.-Forsch. 25 (1975)
77 – 83. 1483 – 1491.
78. J. S. Meyer, T. Sawada, A. Kitamura, M. 99. P. A. van Zwieten, Prog. Pharmacol. 1 (1975)
Toyoda, Neurology (N.Y.) 18 (1968) 772 – 781. 1 – 63.
79. A. Sjöerdsma, A. Vendsalu, K. Engelman, 100. G. Scholtysik, P. Jerie, C. W. Picard in A.
Circulation 28 (1963) 492 – 502. Scriabine (ed.): Pharmacology of
80. L. F. Prescott, R. P. Buhs, J. O. Beattie, O. C. Antihypertensive Drugs, Raven Press, New
Speth et al., Circulation 34 (1966) 308 – 321. York 1980, pp. 79 – 98.
81. S. Z. Langer, Biochem. Pharmacol. 23 (1974) 101. J. C. Doxey, Eur. J. Pharmacol. 54 (1979)
1793 – 1800. 185 – 189.
82. Y. Pommier, M. Andréjak, P. Mouillé, H. 102. A. Jaattela, Eur. J. Clin. Pharmacol. 10 (1976)
Dabiré et al., Naunyn-Schmiedebergs Arch. 73 – 76.
Pharmacol. 318 (1982) 288 – 294. 103. A. J. Bune, J. P. Chalmers, J. R. Graham,
83. J. R. Boissier, J. F. Giudicelli, J. Fichelle, H. P. R. C. Howe et al., Eur. J. Clin. Pharmacol.
Schmitt et al. Eur. J. Pharmacol. 2 (1968) 19 (1981) 309 – 315.
104. I. L. Natoff, B. G. Katzung, F. Weir, J. K.
333 – 339.
Kodama, Pharmacologist 10 (1968) 157.
84. W. Hoefke in A. Scriabine (ed.):
105. T. Baum, Fed. Proc. 28 (1969) 738.
Pharmacology of Antihypertensive Drugs, 106. P. Bolme, H. Corrodi, K. Fuxe, Eur. J.
Raven Press, New York 1980, pp. 55 – 78. Pharmacol. 23 (1973) 175 – 182.
85. H. Schmitt in F. Gross (ed.): Antihypertensive 107. E. Marmo, R. K. Saini, A. P. Caputi, C. Vaca,
Agents, Springer-Verlag, Res. Commun. Chem. Pathol. Pharmacol. 6
Berlin-Heidelberg-New York 1977, (1973) 391 – 406.
pp. 299 – 396. 108. T. Baum, G. Rowles, A. T. Shropshire, Arch.
86. T. Hukuhara, Jr., Y. Otsuka, R. Takeda, F. Int. Pharmacodyn. Ther. 183 (1970) 75 – 84.
Sakai, Arzneim.-Forsch. 18 (1968) 109. P. Bosanac, J. Dubb, B. Walker, M. Goldberg et
1147 – 1153. al., J. Clin. Pharmacol. 16 (1976) 631 – 636.
87. H. Schmitt, H. Schmitt, Eur. J. Pharmacol. 6 110. R. L. Wendt in A. Scriabine (ed.):
(1969) 8 – 12. Pharmacology of Antihypertensive Drugs,
88. W. Hoefke, W. Kobinger, Arzneim.-Forsch. 16 Raven Press, New York 1980, pp. 99 – 111.
(1966) 1038 – 1050. 111. J. W. Strandhoy, B. D. Steg, V. M. Buckalew,
89. G. A. Bentley, I. W. Copeland, J. Starr, Clin. Jr., Life Sci. 27 (1980) 2513 – 2518.
Exp. Pharmacol. Physiol. 4 (1977) 405 – 419. 112. R. H. Meacham, M. Emmett, A. A.
90. M. S. Gold, D. E. Redmond, Jr., H. D. Kleber, Kyriakopoulos, S. T. Chiang et al., Clin.
Lancet 2 (1978) 599 – 602. Pharmacol. Ther. 27 (1980) 44 – 52.
91. R. C. Allen, M. E. Langham, Invest. 113. F. G. McMahon, J. R. Ryan, A. K. Jain, R.
Ophthalmol. 15 (1976) 815 – 823. Vargas et al., Clin. Pharmacol. Ther. 21
92. H. O. Karppanen, E. Westermann, (1977) 272 – 277.
Naunyn-Schmiedebergs Arch. Pharmacol. 279 114. B. R. Walker, M. Geczy, J. A. Gold, Clin.
(1973) 83 – 87. Pharmacol. Ther. 25 (1979) 252.
93. W. Kobinger, Naunyn-Schmiedebergs Arch. 115. L. Dorfman, A. Furlenmeier, C. F. Huebner, R.
Pharmacol. 260 (1968) 153 – 154. Lucas et al., Hel. Chim. Acta 37 (1954)
94. D. S. Davis, E. Neill, J. L. Reid in T. A. Baillie 59 – 75.
(ed.): Stable Isotopes, MacMillan, London 116. R. B. Woodward, F. E. Bader, H. Bickel, A. J.
1978, pp. 45 – 54. Frey et al., J. Am. Chem. Soc. 78 (1956)
95. M. A. Weber, J. Cardiovasc. Pharmacol. 2 2023 – 2025.
117. R. Sannerstedt, J. Conway, Am. Heart J. 79
(1980) Suppl. 1, S 73 – S 89.
(1970) 122 – 127.
96. M. J. M. C. Thoolen, P. B. M. W. M.
118. A. Scriabine in A. Scriabine (ed.):
Timmermans, P. A. van Zwieten,
Pharmacology of Antihypertensive Drugs,
Naunyn-Schmiedebergs Arch. Pharmacol. 319
Raven Press, New York 1980, pp. 119 – 125.
(1982) 82 – 86. 119. M. J. Rand, M. Jurevics in F. Gross (ed.):
97. J. B. Bream, H. Lauener, C. W. Picard, G. Antihypertensive Agents, Springer-Verlag,
Scholtysik et al., Arzneim.-Forsch. 25 (1975) Berlin-Heidelberg-New York 1977,
1477 – 1482. pp. 77 – 159.
26 Antihypertensives

120. R. E. Stitzel, Pharmacol. Rev. 28 (1976) 142. P. Bolli, A. J. Wood, W. L. Phelan, D. R. Lee et
179 – 208. al., Clin. Sci. Mol. Med. 48 (1975)
121. H. Sheppard, R. C. Lucas, W. H. Tsien, Arch. 177 S – 179 S.
Int. Pharmacodyn. Ther. 103 (1955) 143. I. S. Collins, P. Pek, Clin. Exp. Pharmacol.
256 – 269. Physiol. 2 (1975) 445 – 446.
122. R. A. Maxwell, A. J. Plummer, F. Schneider, 144. T. H. Althuis, H.-J. Hess, J. Med. Chem. 20
H. Povalski et al., J. Pharmacol. Exp. Ther. (1977) 146 – 149.
128 (1960) 22 – 29. 145. J. A. Taylor, T. M. Twomey, M. S. von
123. R. A. Maxwell in A. Scriabine (ed.): Wittenau, Xenobiotica. 7 (1977) 357 – 364.
Pharmacology of Antihypertensive Drugs, 146. D. S. Bloom, C. Rosendorff, R. Kramer, Curr.
Raven Press, New York 1980, pp. 127 – 150. Ther. Res. 18 (1975) 144 – 150.
124. R. A. Maxwell, W. B. Wastila in F. Gross (ed.): 147. A. S. P. Hua, I. M. MacDonald, J. B. Meyers, P.
Antihypertensive Agents, Springer-Verlag, Fang et al., Med. J. Aust. 2 (1977) 5 – 9.
Berlin-Heidelberg-New York 1977, 148. W. M. Kirkendall, J. J. Hammond, J. C.
pp. 161 – 261. Thomas, M. L. Overturf et al., JAMA 240
125. B. B. Brodie, C. C. Chang, E. Costa, Br. J. (1978) 2553 – 2556.
Pharmacol. 25 (1965) 171 – 178. 149. Y. K. Seedat, M. A. Seedat, R. Bhoola, S. Afr.
126. F. M. Abboud, J. W. Eckstein, Circ. Res. 11 Med. J. 51 (1977) 461 – 463.
(1962) 788 – 796. 150. A. J. Marshall, D. W. Barritt, J. Pocock, S. T.
127. C. A. Stone, C. C. Porter, J. M. Stavorski, C. T.
Heaton, Lancet 1 (1977) 271 – 274.
Ludden et al., J. Pharmacol. Exp. Ther. 144
151. C. Rosendorff, Br. Med. J. 2 (1976) 508.
(1964) 196 – 204.
152. J. L. Archibald, Chim. Ther. (Paris) 3 (1968)
128. A. N. Brest, JAMA 192 (1965) 41 – 44.
129. J. N. Cohn, T. E. Liptak, E. D. Freis, Circ. Res. 397.
12 (1963) 298 – 307. 153. J. L. Archibald, B. J. Alps, J. F. Cavalla, J. L.
130. H. Villarreal, J. E. Exaire, V. Rubio, H. Dávila, Jackson, J. Med. Chem. 14 (1971)
Am. J. Cardiol. 14 (1964) 633 – 640. 1054 – 1059.
131. C. T. Dollery, D. Emslie-Smith, M. D. Milne, 154. J. L. Archibald in A. Scriabine (ed.):
Lancet 2 (1960) 381 – 387. Pharmacology of Antihypertensive Drugs,
132. F. B. Abramson, C. I. Furst, C. McMartin, R. Raven Press, New York 1980, pp. 161 – 177.
Wade, Biochem. J. 113 (1969) 143 – 156. 155. B. J. Alps, E. T. Borrows, E. S. Johnson, M. W.
133. C. McMartin, Biochem. Pharmacol. 18 (1969) Staniforth et al., Cardiovasc. Res. 6 (1972)
238 – 243. 226 – 234.
134. M. Nickerson, Pharmacol. Rev. 1 (1949) 156. T. Baum, A. T. Shropshire, D. K. Eckfeld, N.
27 – 101. Metz et al., Arch. Int. Pharmacodyn. Ther. 204
135. A. Scriabine, J. W. Constantine, H.-J. Hess, (1973) 390 – 406.
W. K. McShane, Experientia 24 (1968) 157. T. Baum, A. T. Shropshire, Eur. J. Pharmacol.
1150 – 1151. 32 (1975) 30 – 38.
136. J. W. Constantine, W. K. McShane, A. 158. B. J. Alps, M. Hill, K. Fidler, E. S. Johnson et
Scriabine, H.-J. Hess in G. Onesti, K. E. Kim, al., J. Pharm. Pharmacol. 23 (1971)
J. H. Moyer (ed.): Hypertension, Mechanism 678 – 686.
and Management, Grune & Stratton, Inc., 159. B. J. Alps, M. Hill, E. S. Johnson, A. B.
Baltimore 1973, pp. 429 – 444. Wilson, Br. J. Pharmacol. 40 (1970) 153 P.
137. A. Scriabine in A. Scriabine (ed.): 160. G. H. Draffan, P. J. Lewis, J. L. Firmin, T. W.
Pharmacology of Antihypertensive Drugs, Jordan et al., Br. J. Clin. Pharmacol. 3 (1976)
Raven Press, New York 1980, pp. 151 – 160. 489 – 495.
138. I. Cavero, A. G. Roach, Life Sci. 27 (1980) 161. G. H. Draffan, Pharmacol. Clin. 1 (1973)
1525 – 1540. 51 – 59.
139. J. C. Schulz, D. P. Westfall, Blood Vessels 19 162. R. B. Royds, D. J. Coltart, J. D. F. Lockhart,
(1982) 79 – 87. Clin. Pharmacol. Ther. 13 (1972) 380 – 392.
140. G. von der Lippe, O. J. Ohm, P. 163. C. Rosendorff, S. Afr. Med. J. 50 (1976)
Lund-Johansen, Acta Med. Scand. 210 (1981) 764 – 768.
213 – 216. 164. C. T. Dollery, D. G. George, P. J. Lewis, Br. J.
141. J. M. Hayes, R. M. Graham, B. P. O’Connell,
Pharmacol. 46 (1972) 542 P – 543 P.
E. Speers et al., Med. J. Aust. 1 (1976)
562 – 564.
Antihypertensives 27

165. K. Klemm, W. Prüsse, U. Krüger, 186. O. Lederballe Pedersen, Eur. J. Clin.


Arzneim.-Forsch. 27 (1977) 1895 – 1897. Pharmacol. 10 (1976) 381 – 385.
166. W. Schoetensack, P. Bischler, E. C. Dittmann, 187. Å. Hjalmarsson, D. Elinfeldt, J. Herlitz, S.
V. Steinijans, Arzneim.-Forsch. 27 (1977) Holmberg et al. Lancet 2 (1981) 823 – 827.
1908 – 1919. 188. R. J. Lee, D. B. Evans, S. H. Baky, R. J. Laffan,
167. W. Schoetensack, E. G. Bruckschen, K. Zech Eur. J. Pharmacol. 33 (1975) 371 – 382.
in A. Scriabine (ed.): New Drugs Annual: 189. D. B. Evans, M. T. Peschka, R. J. Lee, R. J.
Cardiovascular Drugs, vol. 1, Raven Press, Laffan, Eur. J. Pharmacol. 35 (1976) 17 – 27.
New York 1983, pp. 19 – 48. 190. M. J. Antonaccio, D. B. Evans in A. Scriabine
168. K. Zech in W. Kaufman, E. G. Bruckschen (ed.): Pharmacology of Antihypertensive
(ed.): Proc. 1st Symposium on Urapidil, Drugs, Raven Press, New York 1980,
Excerpta Medica, pp. 295 – 301.
Amsterdam-Oxford-Princeton 1982, 191. R. A. Vukovich, J. E. Foley, B. Brown, D. A.
pp. 50 – 64. Willard et al., Br. J. Clin. Pharmacol. 7 (2)
169. E. G. Bruckschen, F. Henze, G. Michael, (1979) 167 – 172.
Arzneim.-Forsch. 28 (2) (1978) 1176 – 1184. 192. G. Frithz, Curr. Med. Res. Opin. 5 (1978)
170. B. N. C. Prichard, Br. Med. J. 1 (1964) 383 – 387.
1227 – 1228. 193. B. Furberg, A. Dahlqvist, A. Raak, U. Wrege et
171. A. Scriabine, Ann. Rev. Pharmacol. Toxicol. al., Curr. Med. Res. Opin. 5 (1978) 388 – 393.
19 (1979) 269 – 284. 194. R. A. Vukovich, A. Sasahara, P. Zombrano, J.
172. R. Howe, V. A. Cullum, Nature (London) 210 Belko et al., Clin. Pharmacol. Ther. 19 (1976)
(1966) 1336 – 1338. 118.
173. A. M. Barrett, R. G. Shanks, Br. J. Pharmacol. 195. K. Saameli in H. J. Dengler (ed.): Die
34 (1968) 43 – 55. Therapeutische Anwendung β
174. J. D. Fitzgerald in A. Scriabine (ed.): -sympathikolytischer Stoffe, F. K. Schattauer
Pharmacology of Antihypertensive Drugs, Verlag, Stuttgart-New York 1972, pp. 3 – 30.
Raven Press, New York 1980, pp. 195 – 208. 196. C. Weil in A. Scriabine (ed.): Pharmacology of
175. A. S. Nies, D. G. Shand, Circulation 52 Antihypertensive Drugs, Raven Press, New
(1975) 6 – 15. York 1980, pp. 237 – 246.
176. D. G. Shand, E. M. Nuckolls, J. A. Oates, Clin. 197. J. Meier, Curr. Med. Res. Opin. 4 Suppl. 5,
Pharmacol. Ther. 11 (1970) 112 – 120. (1977) 31 – 38.
177. A. Hayes, R. G. Cooper, J. Pharmacol. Exp. 198. R. Gugler, W. Herold, H. J. Dengler, Eur. J.
Ther. 176 (1971) 302 – 311. Clin. Pharmacol. 7 (1974) 17 – 24.
178. T. Walle, E. C. Conradi, U. K. Walle, T. C. 199. M. C. Laver, P. Fang, P. Kincaid-Smith, Med.
Fagan, Clin. Pharmacol. Ther. 27 (1980) J. Aust. 1 (1974) 174 – 176.
22 – 31. 200. A. E. Doyle, Drugs 8 (1974) 422 – 431.
179. L. Hansson, R. Olander, H. Aberg, H.
201. A. S. Turner, J. S. Peel, Med. J. Aust. 2 (1972)
Malmcroma et al., Acta Med. Scand. 190
Spec. Suppl., 48 – 51.
(1971) 531 – 534.
202. J. Rosenthal, H. Kaiser, A. Raschig, D. Welzel,
180. A. P. Douglas-Jones, J. Int. Med. Res. 7 (1979)
Br. J. Clin. Pract. 33 (1979) 165 – 174.
221 – 223.
203. A. Scriabine, M. L. Torchiana, J. M. Stavorski,
181. B. Åblad, K.-O. Borg, E. Carlsson, L. Ek et al.,
C. T. Ludden et al., Arch Int. Pharmacodyn.
Acta Pharmacol. Toxicol. 36 (1975) 7 – 24.
Ther. 205 (1973) 76 – 93.
182. E. Carlsson, B. Åblad, A. Brändström, B.
204. R. A. Hall, R. O. Robson, N. N. Share, Arch.
Carlsson, Life Sci. 11 (1972) 953 – 958.
Int. Pharmacodyn. Ther. 213 (1975),
183. B. Åblad, E. Carlsson, G. Johnsson, C.-G.
pp. 251 – 263.
Regårdh in A. Scriabine (ed.): Pharmacology
205. A. Scriabine in A. Scriabine (ed.):
of Antihypertensive Drugs, Raven Press, New
Pharmacology of Antihypertensive Drugs,
York 1980, pp. 247 –262.
Raven Press, New York 1980, pp. 275 – 282.
184. K.-O. Borg, E. Carlsson, K.-J. Hoffmann, K.-J.
206. D. J. Tocco, A. E. W. Duncan, F. A. De Lauwa
Jönsson et al., Acta Pharmacol. Toxicol. 36
et al., Drug Metab. Dispos. 3 (1975)
(1975) Suppl. 5, 125 – 135.
185. C.-G. Regårdh, G. Johnsson, L. Jordö, L. 361 – 370.
Solvell, Acta Pharmacol. Toxicol. 36 (1975) 207. G. Lohmoller, E. D. Frohlich, Am. Heart J. 89
Suppl. 5, 45 – 58. (1975) 437 – 442.
28 Antihypertensives

208. The Norwegian Multicenter Study Group, N. 230. R. A. Janis, D. J. Triggle, J. Med. Chem. 26
Engl. J. Med. 304 (1981) 801 – 807. (1983) 775 – 785.
209. K. R. H. Wooldridge, Experienta 28 (1972) 231. R. A. Janis, A. Scriabine, Biochem.
1404. Pharmacol. 32 (1983) 3499 – 3507.
210. B. Basil, R. Jordan, A. H. Loveless, D. R. 232. D. J. Triggle in S. F. Flaim, R. Zelis (ed.):
Maxwell, Br. J. Pharmacol. 50 (1974) Calcium Blockers: Mechanisms of Action and
323 – 333. Clinical Applications, Urban &
211. R. H. Briant, C. T. Dollery, C. F. George, Clin. Schwarzenberg, Baltimore 1982,
Trials J. 11 (1974) Suppl. 3, 25 – 28. pp. 121 – 134.
212. B. Levy, J. Pharmacol. Exp. Ther. 186 (1973) 233. R. G. Rahwan, D. T. Witiak, W. M. Muir,
134 – 144. Annu. Rep. Med. Chem. 16 (1981) 257 – 268.
213. B. Basil, R. Jordan in A. Scriabine (ed.): 234. H. Meyer in L. H. Opie (ed.): Calcium
Pharmacology of Antihypertensive Drugs, Antagonists and Cardiovascular Disease,
Raven Press, New York 1980, pp. 283 – 294. Raven Press, New York 1984, pp. 165 – 173.
214. J. P. Amlie, S. Lessum, R. F. Collins, K. 235. H. Meyer, F. Bossert, E. Wehinger, K. Stoepel,
Landmark, Acta Pharmacol. Toxicol. 40 W. Vater, Arzneim.-Forsch. 31 (1981)
(1977) 378 – 388. 407 – 409.
215. D. R. Maxwell, R. F. Collins, Clin. Trials J. 11 236. R. Towart, E. Wehinger, H. Meyer,
(1974) Suppl. 3, 9 – 18. Naunyn-Schmiedebergs Arch. Pharmacol. 317
216. W. L. Ashton, Curr. Med. Res. Opin. 4 (1976) (1981) 183 – 185.
442 – 454. 237. R. Rodenkirchen, R. Bayer, R. Steiner, F.
217. M. Lucsko, J. Guedon, Clin. Trials J. 11 Bossert et al., Naunyn-Schmiedebergs Arch.
(1974) Suppl. 3, 86 – 91. Pharmacol. 310 (1979) 69 – 78.
218. L. Hansson, G. Berglund, O. Andersson, H. 238. M. Hiwatari, K. Satoh, N. Taira,
Holm, Eur. J. Clin. Pharmacol. 12 (1977) Arzneim.-Forsch. 29 (1979) 256 – 260.
89 – 92. 239. B. Garthoff, S. Kazda, Eur. J. Pharmacol. 74
219. B. Carey, E. T. Whalley, J. Pharm. Pharmacol. (1981) 111 – 112.
31 (1979) 791 – 792. 240. K. Aoki, S. Kondo, A. Mochizuki, T. Yoshida
220. J. B. Farmer, I. Kennedy, G. P. Levy, R. J. et al., Am. Heart J. 96 (1978) 218 – 226.
Marshall, Br. J. Pharmacol. 45 (1972) 241. M. T. Olivari, C. Bartorelli, A. Polese, C.
660 – 675. Fiorentini et al., Circulation 59 (1979)
221. G. P. Levy, D. A. Richards in A. Scriabine 1056 – 1062.
(ed.): Pharmacology of Antihypertensive 242. M. D. Guazzi, C. Fiorentini, M. T. Olivari, A.
Drugs, Raven Press, New York 1980, Bartorelli et al., Circulation 61 (1980)
pp. 325 – 347. 913 – 919.
222. D. A. Richards, J. G. Maconochie, R. E. Bland, 243. M. D. Guazzi, A. Polese, C. Fiorentini, A.
R. Hopkins et al., Eur. J. Clin. Pharmacol. 11 Bartorelli et al. Hypertension 5 (1983)
(1977) 85 – 90. Suppl. 2, I 85 – I 90.
244. O. Lederballe Pedersen, N. J. Christensen,
223. L. E. Martin, R. Hopkins, R. E. Bland, Br. J.
K. D. Rämsch, J. Cardiovasc. Pharmacol. 2
Clin. Pharmacol. 3 (1976) Suppl. 1, 695 – 710.
(1980) 357 – 366.
224. B. N. C. Prichard, A. J. Boakes, Br. J. Clin.
245. M. Guazzi, M. T. Olivari, A. Polese, C.
Pharmacol. 3 (1976) Suppl., 743 – 750.
Fiorentini et al., Clin. Pharmacol. Ther. 22
225. P. Bolli, H. J. Waal-Manning, A. J. Wood, F. O.
(1977) 528 – 532.
Simpson, Br. J. Clin. Pharmacol. 3 (Suppl.)
246. O. Lederballe Pedersen, Arch. Int.
(1976) 765 – 771.
Pharmacodyn. Ther. 239 (1979) 208 – 220.
226. H. J. Dargie, C. T. Dollery, J. Daniel, Br. J.
247. M. J. Mulvany, N. Nyborg, Br. J. Pharmacol.
Clin. Pharmacol. 3 (1976) Suppl., 751 – 755.
71 (1980) 585 – 596.
227. A. Fleckenstein, Ann. Rev. Pharmacol.
248. W. Vater, G. Kroneberg, F. Hoffmeister, H.
Toxicol. 17 (1977) 149 – 166.
Saller et al., Arzneim.-Forsch. 22 (1972)
228. T. B. Bolton, Physiol. Rev. 59 (1979)
1 – 14.
606 – 718.
249. B. F. Robinson, R. J. Dobbs, C. R. Kelsey, Br.
229. C. Cauvin, R. Loutzenhiser, C. Van Breemen,
J. Clin. Pharmacol. 10 (1980) 433 – 438.
Ann. Rev. Pharmacol. Toxicol. 23 (1983) 250. S. Kazda, B. Garthoff, G. Luckhaus, Postgrad.
373 – 396. Med. J. 59 (1983) Suppl. 2, 78 – 83.
Antihypertensives 29

251. Y. C. Streifler, C. Wittenberg, J. Rosenfeld in 270. A. Knorr, K. Stoepel, Arzneim.-Forsch. 31


M. Kaltenbach, H. N. Neufeld (ed.): Fifth (1981) 2062 – 2064.
International Adalat Symposium, Excerpta 271. B. F. Johnson, L. Romero, R. Marwaha in A.
Medica, Amsterdam-Oxford-Princeton 1983, Scriabine, S. Vanov, K. Deck (ed.):
pp. 164 – 174. Nitrendipine, Urban & Schwarzenberg,
252. S. Kazda, B. Garthoff, G. Thomas, Drug Baltimore 1984, pp. 535 – 541.
Develop. Res. 2 (1982) 313 – 323. 272. K. Kobayashi, R. C. Tarazi, Hypertension 5
253. S. Kazda, A. Knorr, R. Towart, Prog. (1983) Suppl. 2, I 45 – I 51.
Pharmacol. 5 (1983) 83 – 116. 273. C. E. Hall, S. Hungerford, Clin. Exp.
254. R. A. B. McLeay, T. J. Stallard, R. D. Watson, Hypertens. 5 (1983) 721 – 728.
W. A. Littler, Circulation 67 (1983) 274. D. G. Taylor, S. Shoemaker, T. E. Kowalski in
1084 – 1090. A. Scriabine, S. Vanov, K. Deck (ed.):
255. L.-G. Ekelund, C. Ekelund, S. Rössner, Acta. Nitrendipine, Urban & Schwarzenberg,
Med. Scand. 212 (1982) 71 – 75. Baltimore 1984, pp. 311 – 326.
256. W. Kiowski, O. Bertel, P. Erne, P. Bolli et al., 275. D. C. Warltier, K. A. Lamping, M. G.
Hypertension 5 (1983) Suppl. 1, I 70 – I 74. Zyvoloski, G. J. Gross et al., J. Cardiovasc.
257. A. Zanchetti, in A. Zanchetti, D. M. Krikler Pharmacol. 5 (1983) 272 – 277.
(ed.): Calcium Antagonism in Cardiovascular 276. H. R. Black, N. Vlachakis in A. Scriabine, S.
Therapy: Experience with Verapamil, Vanov, K. Deck (ed.): Nitrendipine, Urban &
Excerpta Medica, Schwarzenberg, Baltimore 1984,
Amsterdam-Oxford-Princeton 1981, pp. 509 – 518.
pp. 292 – 297. 277. J. F. Burris, A. V. Notargiacomo, V.
258. G. Leonetti, C. Cuspidi, L. Sampieri, L. Papademetriou, E. D. Freis, Hypertension 4
Terzoli et al. J. Cardiovasc. Pharmacol. 4 (1982) Suppl. 2, I 32 – I 35.
(1982) Suppl. 3, S 319 –S 324. 278. L. Hansson, L. Andrén, L. Oro, T. Ryman,
259. F. A. Horster in W. Lochner, W. Braasch, G. Hypertension 5 (1983) Suppl. 2, I 25 – I 28.
Kroneberg (ed.): Second International Adalat 279. H. O. Ventura, F. H. Messerli, W. Oigman,
Symposium, Springer-Verlag, New York 1975, F. G. Dunn et al., Am. J. Cardiol. 51 (1983)
pp. 124 – 127. 783 – 786.
260. K. Schlossmann, H. H. Medenwald, H. 280. F. M. Fouad, R. Pedrinelli, E. L. Bravo, F.
Rosenkranz in W. Lochner, W. Braasch, G. Abi-Samra et al., Clin. Pharmacol. Ther. 35
Kroneberg (ed.): Second International Adalat (1984) 768 – 775.
Symposium, Springer-Verlag, New York 1975, 281. H. Haas, E. Busch, Arzneim.-Forsch. 17
pp. 33 – 39. (1967) 257 – 272.
261. S. R. Hamann, R. G. McAllister, J. 282. R. Mannhold, P. Zierden, R. Bayer, R.
Cardiovasc. Pharmacol. 5 (1983) 920 – 927. Rodenkirchen et al., Arzneim.-Forsch. 31
262. C. H. Kleinbloesem, P. van Brummelen, J. A. (1981) 773 – 780.
van de Linde, P. J. Voogd et al., Clin. 283. S. Baky, E. B. Kirsten in M. E. Goldberg (ed.):
Pharmacol. Ther. 35 (1984) 742 – 749. Pharmacological and Biochemical Properties
263. K. D. Rämsch, J. Sommer, Hypertension 5 of Drug Substances, Academy of
(1983) Suppl. 2, I 18 – I 24. Pharmaceutical Sciences, Washington, D.C.
264. R. Krebs, Hypertension 5 (1983) Suppl. 2, 1981, pp. 226 – 261.
I 125 –I 129. 284. S. Baky in A. Scriabine (ed.): New Drugs
265. A. Scriabine, B. Garthoff, S. Kazda, K.
Annual: Cardiovascular Drugs, vol. 2, Raven
Rämsch et al. in A. Scriabine (ed.): New Drugs
Press, New York 1984, pp. 71 – 102.
Annual: Cardiovascular Drugs, vol. 2, Raven
285. B. N. Singh, J. T. Collett, C. Y. Chew, Prog.
Press, New York 1984, pp. 37 – 39.
Cardiovasc. Dis. 22 (1980) 243 – 301.
266. R. Towart, J. Cardiovasc. Pharmacol. 4
286. R. A. Walsh, F. R. Badke, R. A. O’Rourke, Am.
(1982) 895 – 902.
Heart J. 102 (1981) 341 – 350.
267. S. Kazda, B. Garthoff, A. Knorr, Fed. Proc. 42
287. S. F. Flaim, K. Kanda in S. F. Flaim, R. Zelis
(1983) 196 – 200.
(ed.): Calcium Blockers: Mechanisms of
268. H. Meyer, F. Bossert, E. Wehinger, K. Stoepel
Action and Clinical Applications, Urban &
et al., Arzneim.-Forsch. 31 (1981) 407 – 409.
Schwarzenberg, Baltimore 1982,
269. K. Stoepel, A. Heise, S. Kazda,
pp. 155 – 178.
Arzneim.-Forsch. 31 (1981) 2056 – 2061.
30 Antihypertensives

288. B. A. Gould, R. S. Hornung, S. Mann, V. B. 310. T. Meshi, J. Sugihara, Y. Sato, Chem. Pharm.
Subramanian et al., Hypertension 5 (1983) Bull. (Tokyo) 19 (1971) 1546 – 1556.
Suppl. 2, I 91 – I 96. 311. J. E. Francis, ACS Symp. Ser. 27 (1976)
289. K. Midtbo, O. Hals, J. van der Meer, J. 55 – 79.
Cardiovasc. Pharmacol. 4 (1982) Suppl. 3, 312. H. A. J. Struyker-Boudier, H. Van Essen,
S 363 – S 368. J. F. M. Smits, Eur. J. Pharmacol. 95 (1983)
290. P. Erne, P. Bolli, O. Bertel, U. L. Hulthén et al., 151 – 159.
Hypertension 5 (1983) Suppl. 2, I 97 – I 102. 313. E. G. Spokas, H.-H. Wang, J. Pharmacol. Exp.
291. U. L. Hulthen, P. Bolli, F. W. Amann, W. Ther. 212 (1980) 294 – 303.
Kiowski, F. R. Bühler, J. Cardiovasc. 314. I. Khatri, N. Uemura, A. Notargiacomo, E. D.
Pharmacol. 4 (1982) Suppl. 3, S 313 – S 318. Freis, Am. J. Cardiol. 40 (1977) 38 – 42.
292. G. Muiesan, E. Agabiti-Rosei, C. L. Alicandri 315. S. Sen, R. C. Tarazi, Am. J. Physiol. 244
in A. Zanchetti, D. M. Krikler (ed.): Calcium (1983) H 97 – H 101.
Antagonism in Cardiovascular Therapy: 316. M. J. M. C. Thoolen, J. C. A. Van Meel, B.
Experience with Verapamil, Excerpta Medica, Wilffert, P. B. M. W. M. Timmermans et al.,
Amsterdam 1981, pp. 238 – 249. Pharmacology 27 (1983) 245 – 254.
293. A. Chaudhry, M. M. Vohra, Eur. J. Pharmacol. 317. C. Chevillard, M. N. Mathieu, B. Saiag, M.
97 (1984) 156 – 158. Worcel, Br. J. Pharmacol. 69 (1980)
294. G. Haeusler, J. Pharmacol. Exp. Ther. 180 415 – 420.
(1972) 672 – 682. 318. V. S. R. Krishnamurty, Can. J. Physiol.
295. D. G. Taylor, T. E. Kowalski, J. Pharmacol. Pharmacol. 59 (1981) 1159 – 1169.
Exp. Ther. 228 (1984) 491 – 499. 319. M. B. Murphy, A. J. Scriven, M. J. Brown, R.
296. L. H. Opie, Lancet 1 (1980) 806 – 810. Causon et al., Eur. J. Clin. Pharmacol. 23
297. H. Kugita, H. Inoue, M. Ikezaki, M. Konda et (1982) 479 – 482.
al., Chem. Pharm. Bull. (Tokyo) 19 (1971) 320. O. Kohlmann, M. Bresnahan, H. Gavras,
595 – 602. Hypertension 6 (1984) Suppl. 1, I 1 – I 6.
298. H. Kugita, H. Inoue, H. Ikezaki, S. Takeo, 321. E. G. Spokas, G. Folco, J. Quilley, P. Chander,
Chem. Pharm. Bull. (Tokyo) 18 (1970) J. C. McGiff, Hypertension 5 (1983) Suppl. 1,
2028 – 2037. I 107 – I 111.
299. T. Nagao, M. Sato, H. Nakajima, A. Kijomoto, 322. G. Haeusler, M. Gerold,
Jpn. J. Pharmacol. 22 (1972) 1 – 10. Naunyn-Schmiedebergs Arch. Pharmacol. 310
300. M. Sato, T. Nagao, I. Yamaguchi, H. Nakajima (1979) 155 – 167.
et al., Arzneim.-Forsch. 21 (1971) 323. K. Hermsmeyer, A. Trapani, P. W. Abel, M.
1338 – 1343. Worcel, J. Pharmacol. Exp. Ther. 227 (1983)
301. R. P. Hof, Br. J. Pharmacol. 78 (1983) 322 – 326.
375 – 394. 324. C. Chevillard, B. Saiag, M. Worcel, Br. J.
302. H. Narita, T. Nagao, H. Yabana, I. Yamaguchi, Pharmacol. 73 (1981) 811 – 817.
J. Pharmacol. Exp. Ther. 227 (1983) 325. A. J. McLean, P. DuSouish, K. W. Barron,
472 – 477. A. H. Briggs, J. Pharmacol. Exp. Ther. 207
303. M. Sato, S. Murata, H. Narita, M. Tomita et (1978) 40 – 48.
al., Jpn. J. Pharmacol. 75 (1979) 99 – 106. 326. M. Khayyal, F. Gross, V. A. W. Kreye, J.
304. I. Yamaguchi, K. Ikezawa, S. Murata, H. Narita Pharmacol. Exp. Ther. 216 (1981) 390 – 394.
et al., Jpn. J. Pharmacol. 24 (1974) 511 – 522. 327. J. L. Freslon, J. F. Guidicelli, Br. J. Pharmacol.
305. S. F. Flaim in A. Scriabine (ed.): New Drugs 80 (1983) 533 – 543.
Annual: Cardiovascular Drugs, vol. 2, Raven 328. S. Greenberg, J. Pharmacol. Exp. Ther. 215
Press, New York 1984, pp. 123 – 156. (1980) 279 – 286.
306. N. Nakaya, A. Schwartz, R. W. Millard, Circ. 329. L. Hansson, R. Olander, H. Aberg, R.
Res. 52 (1983) 302 – 311. Malmcrona et al., Acta Med. Scand. 190
307. M. Matsuzaki, K. P. Gallagher, J. Patritti, T. (1971) 531 – 534.
Tajimi et al., Circulation 69 (1984) 801 – 814. 330. C. K. Newsome, Southwest Med. 42 (1961)
308. W. Klein, D. Brandt, K. Vrecko, M. Härringer, 558 – 567.
Circ. Res. 52 (1983) Suppl. 1, I 74 – I 173. 331. J. A. Franciosa, G. Pierpont, J. N. Cohn, Ann.
309. M. E. Safar, A. Ch. Simon, J. A. Levenson,
Intern. Med. 86 (1977) 388 – 393.
J. L. Cazor, Circ. Res. 52 (1983) Suppl. 1,
I 169 – I 173.
Antihypertensives 31

332. A. A. Rubin, F. E. Roth, R. M. Taylor, H. 353. C. J. Limas, J. N. Cohn, Circ. Res. 35 (1974)
Rosenkilde, J. Pharmacol. Exp. Ther. 136 609 – 617.
(1962) 344 – 352. 354. G. D. Nicholson, G. A. Alleyne, G. Valdes,
333. T. C. Hamilton, D. Robson, Eur. J. Pharmacol. R. L. Westerman, J. Cardiovasc. Pharmacol. 2
32 (1975) 273 – 278. (1980) Suppl., 242 – 246.
334. W. G. Nayler, I. McInnes, J. B. Swann, D. 355. F. C. Luft, R. Bloch, J. J. Szwed, C. M. Grim et
Race et al. Am. Heart J. 75 (1968) 223 – 232. al., JAMA 240 (1978) 1985 – 1987.
335. P. Limbourg, P. Figel, H. Justin, K. F. Lang, 356. M. Valasco, A. Urbina-Quintana, E.
Eur. J. Clin. Pharmacol. 8 (1975) 387 – 392. Hernández, A. Ramirez et al., Eur. J. Clin.
336. J. Taylor, R. D. Green, Eur. J. Pharmacol. 12 Pharmacol. 20 (1981) 259 – 262.
(1970) 385 – 387. 357. R. E. Mutterperl, F. B. Diamond, D. T.
337. J. C. A. van Meel, P. B. M. W. M. Timmermans, Lowenthal, J. Clin. Pharmacol. 16 (1976)
P. A. van Zwieten, Eur. J. Pharmacol. 92 498 – 509.
(1983) 27 – 34. 358. Y. K. Seedat, R. Rawat, Eur. J. Clin.
338. A. J. Wohl, L. M. Hausler, F. E. Roth, J. Pharmacol. 25 (1983) 9 – 11.
Pharmacol. Exp. Ther. 162 (1968) 109 – 114. 359. E. H. Herman, T. Balazs, R. Young, F. L. Earl
339. N. Winer, D. S. Chokshi, M. S. Yoon, A. D. et al., Toxicol. Appl. Pharmacol. 47 (1979)
Freedman, J. Clin. Endocrinol. 29 (1969) 493 – 503.
1168 – 1175. 360. R. L. Cohen, M. E. Alves, V. C. Weiss, D. P.
340. R. Vandongen, D. M. Greenwood, Eur. J. West et al., J. Invest. Dermatol. 82 (1984)
Pharmacol. 33 (1975) 197 – 200. 90 – 93.
341. G. Sponer, P. Schelling, D. Ganten, F. Gross, 361. V. A. W. Kreye, E. Marquard,
Naunyn-Schmiedebergs Arch. Pharmacol. 303 Naunyn-Schmiedebergs Arch. Pharmacol. 306
(1978) 15 – 20. (1979) 203 – 207.
342. I. I. A. Tabachnick, A. Gulbenkian, F. 362. I. H. Tuzel, J. Clin. Pharmacol. 14 (1974)
Seidman, J. Pharmacol. Exp. Ther. 150 (1965) 494 – 503.
455 – 462. 363. M. Pagani, S. F. Vatner, E. Braunwald,
343. A. McNair, F. Andreasen, P. E. Nielsen, Eur. J. Circulation 57 (1978) 144 – 151.
Clin. Pharmacol. 24 (1983) 151 – 156. 364. R. D. Bastron, G. J. Kaloyanides, J.
344. D. W. DuCharme, W. A. Freyburger, B. E. Pharmacol. Exp. Ther. 181 (1972) 244 – 249.
Graham, R. G. Carlson, J. Pharmacol. Exp. 365. G. Ross, P. V. Cole, Anaesthesia 28 (1973)
Ther. 184 (1973) 662 – 670. 400 – 406.
345. W. A. Pettinger, N. Engl. J. Med. 303 (1980) 366. A. P. Adams, T. N. S. Clarke, J. Edmonds-Seal,
922 – 926. P. Föex et al., Br. J. Anaesth. 46 (1974)
346. D. W. DuCharme, G. R. Zins in A. Scriabine 807 – 817.
(ed.): Pharmacology of Antihypertensive 367. J. Kyncl, Naunyn-Schmiedebergs Arch.
Drugs, Raven Press, New York 1980, Pharmacol. 269 (1971) 390 – 391.
pp. 415 – 421. 368. V. A. W. Kreye in A. Scriabine (ed.):
347. M. Velasco, K. O’Malley, N. W. Robie, J. Pharmacology of Antihypertensive Drugs,
Wells et al., Clin. Pharmacol. Ther. 16 (1974) Raven Press, New York 1980, pp. 373 – 396.
1031 – 1038. 369. C. A. Gruetter, B. K. Barry, D. B. McNamara,
348. H. R. Brunner, P. Jaeger, R. K. Ferguson, E. D. Y. Gruetter, P. J. Kadowitz, L. J. Ignarro, J.
Jequier et al., Br. Med. J. 2 (1978) 385 – 388. Cyclic Nucleotide Res. 5 (1979) 211 – 224.
349. H. N. Sapru, A. J. Krieger, Am. J. Physiol. 236 370. C. A. Gruetter, D. Y. Gruetter, J. E. Lyon, P. J.
(1979) H 174 – H 182. Kadowitz et al., J. Pharmacol. Exp. Ther. 219
350. S. Sen, R. C. Tarazi, F. M. Bumpus, (1981) 181 – 186.
Cardiovasc. Res. 11 (1977) 427 – 433. 371. V. A. W. Kreye, Trends Pharmacol. Sci. 1
351. R. V. Jackson, M. M. Williamson, B. (1980) 384 – 388.
Seneviratne, R. D. Gordon, Aust. N.Z. J. Med. 372. V. A. W. Kreye, G. D. Baron, J. B. Lüth, H.
13 (1983) 39 – 44. Schmidt-Gayk, Naunyn-Schmiedebergs Arch.
352. J. M. McCall, J. W. Aiken, C. G. Chidester, Pharmacol. 288 (1975) 381 – 402.
D. W. DuCharme et al., J. Med. Chem. 26 373. D. H. Foley, Pharmacology 28 (1984)
(1983) 1791 – 1793. 95 – 103.

Você também pode gostar