You are on page 1of 19

Antiulcer Drugs 1

Antiulcer Drugs
This article discusses drugs used in acid-related diseases of the gastrointestinal tract.
Wolfgang Kromer, Byk Gulden, Konstanz, Federal Republic of Germany
Stefan Postius, Byk Gulden, Konstanz, Federal Republic of Germany
Uwe Krüger, Byk Gulden, Konstanz, Federal Republic of Germany

1. Introduction . . . . . . . . . . . . . . . 1 3.4.3. Antimuscarinic Drugs . . . . . . . . . . 12


2. Pathophysiology and Therapeutic 3.4.4. Sucralfate . . . . . . . . . . . . . . . . . . 12
Concepts . . . . . . . . . . . . . . . . . . 1 3.4.5. Bismuth Compounds . . . . . . . . . . 13
3. Drugs . . . . . . . . . . . . . . . . . . . . 2 4. Prospect for Future Developments . 14
3.1. Proton Pump Inhibitors . . . . . . . . 2 4.1. Reversibly Binding Acid Pump An-
3.2. H2 -Receptor Antagonists . . . . . . . 5 tagonists . . . . . . . . . . . . . . . . . . 14
3.3. Antacids . . . . . . . . . . . . . . . . . . 8 4.2. Drugs Directed Against Helicobacter
3.4. Miscellaneous Drugs . . . . . . . . . . 10 pylori . . . . . . . . . . . . . . . . . . . . 14
3.4.1. Prostaglandin Derivates . . . . . . . . . 10 5. Relative Drug Assessment . . . . . . 15
3.4.2. Prokinetic Drugs . . . . . . . . . . . . . 11 6. References . . . . . . . . . . . . . . . . . 15

1. Introduction ter pylori (H.p.), use of nonsteroidal antiinflam-


matory drugs (NSAIDs) such as diclofenac, and
Acid-related diseases of the upper gastrointesti- cigarette smoking. H.p. infection is predomi-
nal tract (GI-tract) comprise all the diseases of nantly acquired in childhood, but the incidence
the esophagus, stomach, and duodenum that can of infections during childhood was considered
be beneficially affected by neutralizing gastric to have decreased over the past decades be-
acid or by inhibiting its secretion. These dis- ing in line with a cohort phenomenon [10],
eases include gastroesophageal reflux disease [11]. Thus, as far as peptic ulcers caused by
(GERD) [1], gastritis [2], gastric and duodenal H.p. infection are concerned, their prevalence
ulcers (GU and DU [3]), drug-induced ulcers [4], was expected to decrease in the future because
Zollinger – Ellison syndrome [5], and stress ul- transmission should become less likely under
cers [6]. However, “acid related” does not mean improved hygienic conditions in industrialized
that all these diseases are causally related to acid countries [12]. On the other hand, Hornemann
hypersecretion. They may in fact go along with et al. [13] found an age-dependent increase in the
normal or even subnormal acid secretion. The prevalence of H.p. infection in healthy German
complex pathophysiology is based on an imbal- children between birth and 17 years of age ad-
ance between aggressive and defensive factors mitted to hospital for minor surgical procedures
and will be discussed in Chapter 2. in 1983 and 1984. Consequently, these authors
Acid-related disorders of the GI-tract still questioned a hygiene-related cohort effect and
constitute a major national-economical issue in interpreted their data as possibly pointing to an
western countries. The “crude lifetime ulcer oral-oral route of infection as opposed to a fecal-
prevalence” has been estimated in the United oral route.
States to be between 8 and 30 % [7]. This prin-
cipal notion also applies to Germany where,
in 1995, 239 × 106 daily doses of antiulcer 2. Pathophysiology and Therapeutic
drugs [H2 -receptor antagonists (H2 -RAs), pro-
ton pump inhibitors (PPIs), etc.] were prescribed Concepts
[8]. Three major risk factors for peptic ulcers and
related disorders have been identified by Ku- Although the dictum “no acid – no ulcer” [14] is
rata and Nogawa [9]: infection by Helicobac- still valid in the majority of peptic ulcer cases,

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


10.1002/14356007.a02 321
2 Antiulcer Drugs

nowadays much more is known about their com- tion 4.2). Figure 1 provides an overview. Finally,
plex etiology and pathophysiology [15]. In fact, the prokinetic drugs cisapride, metoclopramide,
gastric acid and pepsin are the most important and domperidone facilitate propulsive motil-
aggressive factors acting in concert with toxic ity of the GI-tract thus enhancing oral to abo-
factors of H.p. or with either NSAIDs, stress ral movement of contents to prevent duodeno-
or smoking, but they are physiologically coun- gastric reflux of bile acids or gastro-esophageal
terbalanced by defensive factors such as mucus reflux of gastric acid (see Section 3.4.2).
and bicarbonate, epithelial cell lining and re-
pair mechanisms, and mucosal blood flow [15],
[16]. Consequently, although inhibition of acid 3. Drugs
secretion will be therapeutically effective even
in cases with normal acid secretion, eradication This section lays special emphasis on proton
of H.p. by antibiotics or improvement of mu- pump inhibitors as these drugs represent the lat-
cosal defense, for example, by sucralfate and est development in the antisecretory therapy of
antacids, will either eliminate H.p. as the cause peptic ulcer disease, succeeding H2 -receptor an-
of the disease, or shift the balance in favor of tagonists. In general, information on chemical
defensive factors. Bismuth compounds do a lit- and pharmaceutical drug properties, synonyms,
tle of both. Prostaglandin (PG) derivatives help trade names, dosages etc. beyond the informa-
preventing NSAID-induced ulcers by “gastro- tion given below may be obtained from [22–26].
protective” (misleadingly also called “cytopro- In the following sections, the chemical names of
tective”) mechanisms but heal peptic ulcers only individual drugs are indicated according to the
when given in antisecretory doses [17], [18]. Ul- WHO nomenclature.
cer pain may be triggered by exposure of the
inflamed mucosa to gastric acid [19] and will
therefore be alleviated by antisecretory or acid- 3.1. Proton Pump Inhibitors
neutralizing drugs, apart from their healing ef-
fect. Chemically, proton pump inhibitors (PPIs) con-
The most effective target of acid-inhibitory sist of a substituted benzimidazole moiety linked
drugs is the H+ /K+ -ATPase (acid pump; pro- by an essential sulfinylmethyl group to a sub-
ton pump) of the parietal cell’s apical mem- stituted pyridine ring [20], [27]. They are acid-
brane [20]. PPIs (pantoprazole, omeprazole, lan- labile prodrugs that have first to be activated by
soprazole, see Section 3.1) block the final step acid in order to gain pharmacological activity but
in acid production (Fig. 1) and therefore dis- will subsequently be degraded to inactive prod-
play a higher antisecretory efficacy and clin- ucts (see below). They are therefore adminis-
ical reliability than H2 -RAs (see Section 3.2) tered as enteric-coated formulations (which pro-
or, even more so, M1 -antimuscarinics. H2 - tects the prodrug from acid in the gastric lumen),
RAs block receptors for histamine located up- become absorbed from the intestine and pene-
stream on the basolateral membrane of the trate into the acid compartment of the gastric
parietal cell (H2 -receptor), M1 -antimuscarinics parietal cell from the blood. The pH in the order
block receptors for actetylcholine on either of 1 present inside the canaliculus of a secreting
the histamine-liberating ECL-cell or intramural parietal cell [20] causes protonation of the pyri-
neurons (M1 -receptors). Consequently, both of dine nitrogen (pK a about 4) and thereby drug
these leave other pathways of acid stimulation accumulation by a factor of more than 1000 rel-
unaffected. The only antimuscarinic antiulcer ative to the blood. This protonation goes along
drug still in use is pirenzepine (see Section 3.4.3) with a complex, intramolecular rearrangement
which seems outdated due to its comparatively of the prodrug structure to form a thiophilic
low efficacy combined with unwanted effects. cyclic sulfenamide. The latter is a highly reac-
Antacids (see Section 3.3), PG derivatives (see tive compound and would be further degraded
Section 3.4.1), sucralfate (see Section 3.4.4) and to inactive products (see above) if it were not
bismuth compounds (see Section 3.4.5) display generated in the vicinity of the extracytoplas-
complex actions partially related to acid, mu- mic portion of the proton pump where it imme-
cosal defensive mechanisms, and H.p. (see Sec- diately and covalently reacts with thiol groups of
Antiulcer Drugs 3

Figure 1. Simplified and incomplete illustration of the interplay between various aggressive and defensive gastric mucosal
factors, their physiological regulation, and the different drug targets in peptic ulcer therapy.
ACh = acetylcholine; ECL = enterochromaffine-like; G = gastrin receptor; Hist = histamine; H2 = histamine H2 -receptor;
M1 = muscarinic M1 -receptor; N = nicotinic receptor; NSAIDs = nonsteroidal antiinflammatory drugs; PG = prostaglandin
receptor.
X indicates inhibition of the respective target.
After Brunton [21], slightly modified.

the proton pump. The result is a mixed disulfide administration, and any co-administration of re-
of the active drug and the enzyme protein [28], ceptor antagonists (be it H2 - or M1 -blockers)
[29]. The disulfide structure arrests the catalytic with PPIs is contraindicated. In fact, the latter
cycle of the proton pump [20] and thus inhibits would lose instead of gain efficacy because re-
acid secretion. Although this constitutes an irre- ceptor blockade would prevent a certain percent-
versible mode of inhibition in a chemical sense, age of parietal cells from being stimulated for
acid inhibition by PPIs is biologically reversible acid secretion thus kept in a resting state not al-
due to the turnover of both the proton pump (t 1/2 lowing for PPI activation.
approx. 72 or 54 h for the rat proton pump or its Cyclic sulfenamides generated by acid from
alpha subunit; [30], [31]) and the parietal cell the inactive prodrugs (for review, see [27]) are
(turnover time in the rat ca. 164 d [32]). specific SH-reagents which can modify any thiol
It has been estimated that about one-third of group and should therefore be prevented from
the parietal cell mass is in its resting state even unwanted reactions at any targets apart from the
during meal stimulation [33] and therefore not parietal cell’s proton pump in order to avoid
available for inhibition by PPIs [34]. These rest- side effects as much as possible. Thus, the main
ing cells are liable to endogenous stimulation af- aspect governing selection of candidate com-
ter the drug has been eliminated from blood cir- pounds during drug development was their pH-
culation and thus mainly determine the recovery dependent activation profile. As discussed in de-
of acid secretion following a first drug admin- tail by Kromer et al. [37], PPIs should be acid-
istration. It also follows that PPIs achieve their activated as fast as possible at low pH values
steady state effect only over about 3 days of daily of about 1 in order to potently inhibit the pari-
4 Antiulcer Drugs

etal cell’s proton pump. However, they should interactions [57]. By contrast, phase II conjuga-
be activated as slowly as possible at pH values tion is lacking in case of omeprazole, which in
well above 3 to avoid unwanted SH-reactions addition has an about seven-fold higher affin-
in moderately acidic tissue compartments like ity to human cytochrome P450 than pantopra-
lysosomes and other acidic vesicles. “Fast” and zole [58]. The two differences, i.e., lower affin-
“slow” activation is always meant relative to ity to cytochrome P450 and phase II conjugation
the serum elimination half-life t 1/2 . For exam- of pantoprazole as opposed to omeprazole ex-
ple, omeprazole is as quickly activated at pH plain well that pantoprazole did not display any
5 (where activation is unwanted) as it is elim- metabolic drug interactions in man [59], [60],
inated from serum (t 1/2 approx. 1 h), whereas whereas omeprazole did in a number of studies
the PPI pantoprazole is eliminated from blood [61–70], for review, see [71].
by a factor of about 3 faster than it is activated Another goal considered during PPI selection
at pH values in the order of 5 [37]. For lanso- and development was to obtain linear pharma-
prazole and rabeprazole, see [35], [36]. All of cokinetics both with respect to dose increments
these drugs are acid-activated at pH 1 with ac- and repeated dosing over time. This goal has
tivation half-lifes in the order of a few minutes been achieved with pantoprazole [72–74] but not
compared to their serum elimination half-lifes with omeprazole [75].
of about 1 hour. They all display therefore sim- All of the differences in the aforementioned
ilar antisecretory potencies and efficacies [38– drug properties depend on the different sub-
44] which solely depend on the activation rate stitution patterns of the two ring systems of
within the acid space of the parietal cell. For the the PPIs (Table 1). Regarding the pH-selectivity
same reason, 20 mg omeprazole achieved lower in the PPI’s chemical activation (see above),
healing rates than 40 mg of either omeprazole “the difluoromethoxy group present in panto-
or pantoprazole in a number of studies [45–51]. prazole slows reaction at neutral pH by elec-
This data does not conflict at all with the failure tron withdrawal, which decreases the pK a of
of other studies [52], [53] to statistically distin- the benzimidazole N and reduces its protona-
guish 40 mg pantoprazole from 20 mg omepra- tion at all but highly acidic pH values” [20].
zole since the two doses represent the upper The higher lipophilicity of omeprazole explains
part of the dose – response relationship. Conse- its higher volume of distribution (i.e., a higher
quently, they are separated by only a small (but percentage shifted from serum into tissues)
clinically relevant) difference in healing rates and, consequently, its somewhat lower maxi-
confounded by comparatively large variations mum serum concentration (C max ) and area un-
between different patient samples. It has there- der the serum concentration – time curve (AUC)
fore been judged from a medical rather than a (→ Pharmaceutical Dosage Forms, Chap. 1. on
statistical point of view that 20 mg omeprazole bioavailability) compared to the same dose of
daily being underdosed and 40 mg daily of both pantoprazole [37], [76]. The biological implica-
omeprazole and pantoprazole being the correct tions of these differences have been discussed
and desirable dose for acute ulcer healing [54], in [37]. The individual PPIs will be presented
[55]. Figure 2 demonstrates the congruence be- below in alphabetical order.
tween animal data, human pharmacology data,
Table 1. Chemical structures of four proton pump inhibitors
and selected clinical data (see above) and shows
the equipotency and equiefficacy of pantopra-
zole compared to omeprazole on a milligram
basis.
A further goal pursued during selection of
PPIs was to minimize potential interactions with Lansoprazole
cytochrome P450 in order to avoid unwanted
drug interactions in the liver. Following phase
I oxidation in the endoplasmic reticulum of
the liver cell, pantoprazole undergoes cytosolic
phase II sulfate conjugation which is indepen- Omeprazole
dent of cytochrome P450 and not subject to drug
Antiulcer Drugs 5

logical doses [84], gastric bacterial overgrowth


due to anacidity [85], impaired absorption of
vitamin B12 and iron [86], induction of cy-
tochrome P450 IA2 which both generates and
detoxifies mutagens [87], [88]; suspected geno-
Pantoprazole toxicity [89], and very rare events not conclu-
sively related to omeprazole treatment (impaired
vision and hearing, inflammatory and immuno-
logic events like fever, vasculitis and colitis).
All of these issues have been reviewed by ex-
Pariprazole/Rabeprazole/E3810 perts and considered to be of either no rele-
vance or even unrelated to omeprazole treat-
ment [86], [90–96]. The aforementioned adverse
Lansoprazole [103577-45-3], 2-[[[3- events published as case reports seem to be ex-
methyl-4-(2,2,2-trifluoroethoxy)-2-pyridyl]- tremely rare relative to the large number of treat-
methyl]sulfinyl]benzimidazole (WHO), ment courses with omeprazole of well above
C16 H14 F3 N3 O2 S, M r 369.37 [77], available 100 million. Some of these adverse effects, how-
as capsules containing either 15 or 30 mg (free ever, disappeared upon cessation of omepra-
acid) of the racemic mixture (chiral center at the zole therapy and reappeared upon rechallenge
asymmetric sulfinyl group). Both the (+)- and suggesting a causal relationship. These include
(−)-enantiomers of lansoprazole are converted cases of myopathy [97], neuromyositis [98], ery-
to the optically inactive cyclic sulfenamide and thema nodosum [99], arthralgia [100], pityriasis
are therefore of the same antisecretory potency rosea-like skin eruption [101], hemolytic anemia
in vitro [78]. [102], interstitial nephritis [103], [104], CNS-
Trade names: Agopton (Takeda) and in ad- symptoms [105], [106], and acute gout [107].
dition various licensed products under different
trade names (see [23]). Pantoprazole [102625-70-7], 5-(difluoro-
methoxy)-2-[[(3,4-dimethoxy-2-pyridyl)methyl]-
Omeprazole [73590-58-6], 5-methoxy-2- sulfinyl]benzimidazole (WHO), sodium salt
[[(4-methoxy-3,5-dimethyl-2-pyridyl)methyl]- C16 H15 F2 N3 NaO4 S, M r 405.38, [27], [53],
sufinyl]benzimidazole (WHO), C17 H19 N3 O3 S, [56], [108–111]. Figure 3 shows the synthe-
M r 345.42 [52], available as capsules contain- sis of the sodium salt, Figure 4 its chemical
ing either 10, 20, or 40 mg and as a lyophylisate activation. Pantoprazole is available as enteric
for i.v. infusion containing 40 mg of the racemic coated tablets containing 45.1 mg of the sodium
mixture (40 mg being equivalent to 42.6 mg of salt sesquihydrate equivalent to 40 mg of the
the sodium salt). The omeprazole enantiomers racemic mixture of the free anhydrous acid,
can be separated but both of them display about and as a lyophylisate for i.v. infusion. The (+)-
the same antisecretory potency in vitro [79], and (−)-enantiomers do not differ in their anti-
[80]. secretory potencies in animals. An oral 20 mg
Trade names: The racemic mixture is pro- formulation for GERD/maintenance therapy has
duced by Astra/Hässle under trade names Antra been approved recently.
and Losec, and is in addition distributed by Trade names: Pantozol (Byk Gulden) and
various licensees under different trade names others; various licensed products under differ-
(see [23]). ent trade names (see [23]).
Omeprazole was the first PPI in clinical use
and has therefore attained the largest drug safety
data base. Omeprazole has a remarkable safety 3.2. H2 -Receptor Antagonists
profile with most adverse events in the range of
that of placebo treatment [81–83]. Challenges H2 -receptor antagonists (H2 -RAs) are structural
had been hypergastrinemia (also seen with the analogues of histamine that – at least those in
other PPIs) with subsequent induction of carci- clinical use – competitively and surmountably
noids in rats chronically exposed to high toxico-
6 Antiulcer Drugs

Figure 2. Congruence between animal data, human pharmacology data and clinical data with respect to equipotency and
equiefficacy of two gastric proton pump inhibitors.A) Inhibition of gastric acid secretion in various animal models: Inhibitory
doses, ID50 values (µmol/kg; 1 µmol = 0.35 mg of omeprazole or 0.38 mg of pantoprazole, free acid). HPD = Heidenhain
pouch dog, FR = acute fistula rat, GSR = Ghosh-Schild rat, 2-DG = 2-deoxy-d-glucose. The routes of administration refer to
test drugs, not secretagogues (impromidine, penta. = pentagastrin, bethan. = bethanechol). For further information, see [56].B)
24 h-pH-metry in healthy volunteers (time of intragastric pH > 4). All doses 40 mg orally, daily over 7 d Left pair of columns:
Koop et al. [40]. Right pair of columns: Brunner et al. [44]. The comparison between the two studies shows that, although PPIs
may vary in their efficacy depending on the random sample, omeprazole and pantoprazole proved equipotent when compared
intraindividually.C) Healing rates in gastroesophageal reflux disease (GERD), duodenal ulcer (DU), and gastric ulcer (GU) in
a comparison between daily oral doses of either 20 or 40 mg omeprazole or 40 mg pantoprazole. Selected studies that show a
clear cut advantage of the 40 mg dose. From left to right: Hetzel et al. [45]; Laursen et al. [46]; Bate et al. [47]; Lauritsen
et al. [48]; Walan et al. [49]; Valenzuela et al. [50]; Witzel et al. [51]. The specifications “all” and “> 1 cm” below the
sixth group of columns [50] refer to the ulcers evaluated. p-Values are indicated at the bottom of columns compared to each
other. All of the differences between the two doses shown in the figure are statistically significant irrespective of the particular
drug involved. It is important to note that the failure of other studies (not shown here; see [50] and [53]) to demonstrate any
significant difference between the healing rates upon 40 mg pantoprazole compared to 20 mg omeprazole does not conflict
with the aforementioned superiority of the 40 mg dosage. In fact, 40 mg of either drug represent a ceiling dose separated by
a comparatively small difference from the healing rate upon 20 mg. Thus, both of the two doses are localized in the upper
portion of the dose response curve. Because of the large interindividual variation seen in clinical studies, the difference will
be statistically significant in one, but statistically insignificant in another study.
Antiulcer Drugs 7

block the H2 -receptors located on the basolat-


eral membrane of the parietal cell. Histamine is
the major secretagogue in humans [33], [112].
An imidazole ring as part of the structure was
originally regarded essential for specific H2 -
antagonism but this was refuted by the newer
H2 -RAs (see [113]). They show a similarly fa-
vorable safety profile as the PPIs [114] but face
the problem of about 10 % resistant peptic ulcers
[115] and loss of effectiveness by about 50 %
over a 4-week treatment course (tolerance devel-
opment [116]). Potential reasons have been dis-
cussed in [117], [33]. Table 2 shows the chemical
structures of the H2 -RAs. In contrast to cime-
tidine (imidazole ring), the newer compounds
contain thiazole (famotidine and nizatidine), fu-
ran (ranitidine) or piperidine and benzene (rox-
atidine) ring systems. This helps to avoid un-
wanted cytochrome P450, i.e., metabolic drug
interactions [118]. The individual drugs are dis-
cussed in the following in alphabetical order.

Table 2. Chemical structures of H2 -receptor antagonists. Note the


different ring systems. The imidazole ring is mainly responsible for
metabolic drug interactions [118].

Cimetidine

Famotidine

Figure 3. Synthesis of pantoprazole sodium as an example


of PPIs [108]
Nizatidine

Cimetidine [51481-61-9], 1-cyano-2-


methyl-3-[2-[[(5-methylimidazol-4-yl)methyl]-
thio]ethyl]guanidine (WHO) [119],
C10 H16 N6 S, M r 252.34, mp 141 – 143 ◦ C, sol-
Ranitidine
ubility in water at 37 ◦ C 1.14 wt % increased by
dilute HCl, pK a -values 6.8 and 7.1. Cimetidine
is a crystalline powder with bitter taste and char-
acteristic odor. Citmetidine was the first of the
Roxatidine H2 -RAs and revolutionized peptic ulcer therapy
when introduced in the 1970s [120].
Trade names: Tagamet (SmithKline
Beecham) and numerous others (see [23]).
8 Antiulcer Drugs

69 – 70 ◦ C, freely soluble in water. Ranitidine


was the second H2 -receptor antagonist intro-
duced in the early 1980s. It further optimized
peptic ulcer therapy by minimizing unwanted
interactions with androgen receptors and cy-
tochrome P450 (see above). It has a higher
affinity and, hence, greater potency compared to
cimetidine which, however, is important only in
the context of the relative drug safety margins.
Trade names: Zantic (Glaxo) and numerous
others (see [23]). Ranitidine is also available as
Raniditine bismuth citrate used in H.p.-positive
ulcer patients (see Section 3.4.5).

Roxatidine [78273-80-0], N-[3-[(α-


piperidino-m-tolyl)oxy]-propyl]glycolamide
(WHO) [124], C17 H26 N2 O3 , M r 306.41, mp
59 – 60 ◦ C.
Trade name: Roxit (Albert-Roussel).

3.3. Antacids

Starting with naturally occurring carbonates


centuries ago and later followed by potash com-
Figure 4. The chemical activation pathway of substituted
bined with bismuth more than one century ago,
benzimidazoles exemplified by pantoprazole (according to antacids have since been developed and widely
[27])E = enzyme used over decades (see [125]). They are better
than placebos and as effective as cimetidine in
DU healing but less effective in GU healing
Famotidine [76824-35-6], [1-amino-3- [125]. Available both as liquids and tablets, they
[[[2-[(diaminomethylene)-amino]-4-thiazolyl]- are nowadays usually taken 1 h after each meal
methyl]thio]propylidene]-sulfamide (WHO) and at bedtime in a total daily dose equivalent
[121], C8 H15 N7 O2 S3 , M r 337.43, mp to about 400 mmol neutralizing capacity. This
163 – 164 ◦ C, solubility in water at 20 ◦ C should be sufficient for all patients while min-
0.1 wt %. imizing adverse effects (see below). Such regi-
Trade names: Pepdul (Merck Sharp & mens elevate intragastric pH above 3.5 for about
Dohme) and numerous others (see [23]). 2 h. Water-insoluble, aluminum – magnesium-
containing antacids (like magaldrate) leave the
Nizatidine [76963-41-2], N-[2-[[[2-[(di- stomach more slowly with the solid meal por-
methylamino)methyl]4-thiazolyl]methyl]- tion and are therefore longer acting. In ad-
thio]ethyl]N’-methyl-2-nitro-1,1-ethenediamine dition to acid neutralization, these aluminum-
(WHO) [122], C12 H21 N5 O2 S2 , M r 331.45, mp containing antacids enhance the production
130 – 132 ◦ C, solubility in water 0.1 – 0.33 wt %, of mucosal prostaglandins and possibly SH-
pK a -values 2.1 and 6.8. containing compounds to protect the mucosa
Trade names: Nizax (Lilly) and others from injury (“gastroprotection”) [125]. In ther-
(see [23]). apy and prevention of mild GERD, acid neu-
tralization, subsequent partial pepsin inactiva-
Ranitidine [66357-35-5], N-[2-[[5-[(di- tion, and binding of bile acids and lysolecithin
methylamino)-methyl]furfuryl]thio]ethyl]N’- may act in concert (see [125]). Antacids display
methyl-2-nitro-1,1-ethenediamine (WHO) a favorable cost : benefit ratio in mild diseases.
[123], C13 H22 N4 O3 S, M r 314.41, mp
Antiulcer Drugs 9

More severe cases of acid related disorders re- For tabulated overviews of antacids, see [21],
quire PPIs (see Section 3.1). [131], [132]. The indicated in vitro neutraliza-
Although antacids did not display any ter- tion capacities do not necessarily reflect the rel-
atogenic effects in animals and may therefore ative therapeutic efficacies which are also af-
be used in pregnancy- and delivery-associated fected by pH-dependent dissolution, viscosity,
GERD (see [125]), they can interfere with iron gastroprotective effects, etc. (see magaldrate).
absorption and may facilitate metabolic alcalo- Antacid formulations may contain many addi-
sis and fluid overload. The latter effects depend tives such as simethicone (carminative contained
on their bicarbonate and sodium ion content in Riopan; see [131]); these are not considered
which is considered obsolete. In general, low here. Only the major antacid components which
sodium products should be preferred. Particulate influence gastric acidity will be discussed below
antacids may also cause pneumonia upon aspi- in alphabetical order.
ration and have been substituted for prevention
of reflux complications largely by antisecreta- Aluminum hydroxide [21645-51-2],
gogues. Nonulcer dyspepsia (NUD) and prophy- Al(OH)3 , M r 77.99, insoluble in water, solu-
laxis of NSAID-induced ulcers (see misoprostol, ble in dilute mineral acids and alkali hydroxides
Section 3.4.1) are no recommended indications (4 % suspension in water), white, amorphous
for antacids. Apart from the aforementioned ad- powder. For other relevant properties and pro-
verse effects, aluminum-containing antacids can duction, see → Aluminum Oxide, Chap. 1.1.
bind tetracyclines and many other drugs and Available as Al(OH)3 ·nH2 O (algedrate) and
thus prevent their absorption [21]. Aluminum- algedrate hexitol complex. Depending on gastric
containing antacids may also complex phos- acidity, stepwise buffering proceeds according
phate (to cause osteomalacia in the worst case) to
and constipate the patient. The latter effect is
Al(OH)3 + 3 HCl  Al(OH)2 Cl + H2 O + 2 HCl
counterbalanced by magnesium [21] contained
 Al(OH)Cl2 + 2 H2 O + HCl  AlCl3 + 3 H2 O
in combination formulations such as magal-
drate (see [126]), because magnesium salts in Pure Al(OH)3 ·nH2 O is not very stable, it ages
the intestine increase GI motility and cause os- at room temperature upon loss of water (forma-
motic diarrhea [127]. HCl converts insoluble tion of aluminum oxide) and neutralizing capac-
aluminum oxide and hydroxide into more read- ity. Aluminum hydroxide is preferably used as
ily soluble and absorbable aluminum chloride. a combination preparation with magnesium hy-
However, the causal relationship between alu- droxide to counterbalance gut motility effects.
minum intake with antacids and Alzheimer’s This mixture also displays increased stability.
disease is unlikely, while chronic intake of high Generated from aluminum hydroxide and gas-
doses may cause CNS toxicity in patients with tric acid, aluminum chloride binds phosphate as
renal insufficiency [128], [129]. insoluble AlPO4 which is then excreted with
Antacids containing carbonate, namely feces. Aluminum hydroxide is therefore also
NaHCO3 or CaCO3 , produce CO2 with HCl used at mealtime in patients on dialysis to re-
and cause therefore gastric flatulence, belching duce phosphate intake. Magnesium containing
and acid reflux. Na+ may cause problems in hy- antacids (see below) are contraindicated in this
pertensive patients, Ca2+ adversely stimulates condition to avoid dangerous hypermagnesemia
gastrin and HCl secretion (“acid rebound”). [132].
Those salts of carbonic acid are considered ob- Trade names: Actal and numerous others
solete even in antacid mixtures although they (see [23]).
are still used to some extent. Modern alterna-
tives are the poorly soluble bases magnesium Aluminum phosphate [7784-30-7],
and aluminum hydroxide. If the two are incor- AlPO4 , M r 121.95, white powder, insoluble in
porated together into one tridimensional lattice water, soluble in concentrated mineral acids, pH
as represented by magaldrate (see below), they 5.5 – 6.5 (4 % suspension in water). Available as
behave as a monosubstance buffering the pH AlPO4 gel or AlPO4 ·2 H2 O. For other relevant
between about 3 and 5 [130]. properties and production, see → Phosphoric
Acid and Phosphates, Chap. 3.3.6.
10 Antiulcer Drugs

Trade names: Phosphalugel (Boehringer In- that of other antacids, its in vivo neutralizing ca-
gelheim) and others (see [23]). pacity in humans is significantly higher [142]
due to the exclusion of proteins [133]. How-
Magnesium hydroxide [1309-42-8], ever, magaldrate forms a gel with a large inner
Mg(OH)2 , M r 58.32, amorphous powder, vir- surface confining water spaces readily accessi-
tually insoluble in water (pH 9.5 – 10.5), solu- ble to HCl. The alternation of magnesium and
ble in dilute acids. For other relevant properties aluminum hydroxides within the layered lattice
and production, see → Magnesium Compounds, prevents losses of buffering capacity by aging.
Chap. 4. Available in combination preparations Magaldrate thereby shows prolonged acid neu-
with aluminum hydroxide and under numerous tralization up to the optimum of pH 4 [133]. In
trade names (see [23]). The equation for neu- contrast, an aging phenomenon is found in alu-
tralization is minum hydroxide gels which is due to transition
of aluminum hydroxide to aluminum oxide upon
Mg(OH)2 + 2 HCl  MgCl2 + 2 H2 O
loss of water.
For potential side effects, see general discussion The magaldrate gel binds cholic acids like an
of antacids. anion exchanger, and it binds lysolecithin and
pepsin on its outer surface [143]. It provides bal-
Magaldrate (hydroxymagnesium alumi- anced, opposing actions of aluminum and mag-
nate) [74978-16-8], Al5 Mg10 (OH)31 (SO4 )2 ·nH2 O, nesium on gut motility [21].
insoluble in water, white, odorless crystalline
powder, soluble in dilute mineral acids. Mag-
aldrate [133] favorably combines the strongly 3.4. Miscellaneous Drugs
basic properties of magnesium located in the
crystal core with the less basic properties of alu- 3.4.1. Prostaglandin Derivates
minum ions surrounding the former in a layered (→ Prostaglandins)
lattice (Fig. 5). The magaldrate layered lattice
consists of recurrent stacks of octahedral ba- Prostaglandin (PG) derivatives exert gastropro-
sic layers with an excess of positive charges tective effects at subantisecretory doses [144]
[Al5 Mg10 (OH)30 ]5+ on the outer gel surface but have the disadvantage of frequent side ef-
[134], [135]. These charges electrostatically in- fects (mostly diarrhea and cramps) when ad-
teract with negative charges of the gastric mucus ministered at higher, antisecretory doses [128].
layer to favorably increase the antacid’s reten- Only the latter accelerate healing of GU and DU.
tion time in the stomach [136]. Prostaglandins seem to be somewhat inferior to
Trade names: Riopan (Byk Gulden) and nu- H2 -receptor antagonists [145], particularly with
merous others (see [23]). respect to pain relief. Prostaglandins are abor-
In these brucite-like layers, about one third of tifacient and are contraindicated during preg-
the HO – Mg – OH units are substituted by pos- nancy and lactation. Low doses of PG deriva-
itively charged HO – Al+ – OH units, counter- tives have proved valuable in the prophylaxis of
balanced by SO2− −
4 and OH anions in the aque- NSAID-induced peptic ulcers [21], [132]. Apart
ous interlayer [137–139]. Compared to antacids from the PG-E1 derivative misoprostol (see be-
composed of mixed hydroxides of aluminum low), other PG-E1 (rioprostil) and PG-E2 deriva-
and magnesium, magaldrate has a low aluminum tives (arboprostil, enprostil, trimoprostil) had
content per milliliter and releases less aluminum been under study [21], [146].
because it rebinds aluminum ions in exchange
for the more basic magnesium ions provided by Misoprostol [59122-46-2], 15-deoxy-
an excess of antacid [140], [141]. A further ad- 16-hydroxy-16-methyl-PG-E1 , (±)-Methyl-
vantage of the layered lattice is that its inner (1R,2R,3R)-3-hydroxy-2-[(E)-(4RS)-4-
surface is not accessible to aluminum-binding hydroxy-4-methyl-1-octenyl]-5-oxocyclopen-
proteins that otherwise reduce the buffering taneheptanoate (WHO) [147], C22 H38 O5 , M r
capacity of aluminum-containing conventional 382.54, light yellow oil, slightly soluble in wa-
antacids [142]. In fact, although the in vitro acid- ter. Misoprostol is approved for prophylaxis of
neutralizing capacity of magaldrate is similar to
Antiulcer Drugs 11

Figure 5. Schematic drawing of the layered lattice of magaldrate, a special example of an antacid (according to [134])

NSAID-induced ulcers and the treatment of DU diarrhea; [152]). Domperidone has the advan-
and GU regardless of the limitations mentioned tage over metoclopramide of not significantly
above. entering the central nervous system (CNS), thus
Trade names: Cytotec (Heumann) and others avoiding central side effects. Although cisapride
(see [23]). concentrations in rat brain can reach one-third
to half of the plasma concentration, it does
not block dopamine receptors and is therefore
3.4.2. Prokinetic Drugs devoid of extrapyramidal motor and prolaktin
Reflux esophagitis (GERD) is caused by reflux effects [152]. Drug interactions by prokinetic
of gastric acid and therefore treated not only by drugs have been reviewed in [153].
PPIs and antacids but also by prokinetic drugs
which facilitate oral to aboral clearing of the Cisapride [81098-60-4], cis-4-amino-5-
esophagus and emptying of the stomach [148]. chloro-N-[1-[3-(p-fluorophenoxy)-propyl]-3-
Prokinetic drugs do so by alternately decreasing methoxy-4-piperidyl]-o-anisamide (WHO)
the frequency of intermittent lower esophageal [152], C23 H29 ClFN3 O4 , M r 465.95, available
sphincter relaxations, and increasing the ampli- as monohydrate, mp 109.8 ◦ C is, like metoclo-
tude and frequency of esophageal as well as gas- pramide, a substituted benzamide.
tric peristaltic contractions. Two pharmacologi-
cal principles are used: (1) blockade of periph-
eral dopamine D2 -receptors by domperidone or
metoclopramide which may lead to disinhibi-
tion of motility (although its mediation by D2 -
receptor blockade is controversial), and (2) ac-
tivation of peripheral 5-HT4 -receptors by either
metoclopramide or cisapride. The latter inter- Trade names: Propulsin (Janssen) and others
action triggers the release of acetylcholine that (see [23]).
subsequently stimulates motor neurons [149],
[150]. Side effects relate primarily to either Domperidone [57808-66-9], 5-chloro-1-
blockade of dopamine receptors (extrapyrami- [1-[3-(2-oxo-1-benzimidazolinyl)propyl]-4-
dal motor and prolaktin effects as well as diar- piperidyl]-2-benzimidazolinone (WHO) [154],
rhea; [151]) or release of acetylcholine (cramps,
12 Antiulcer Drugs

C22 H24 ClN5 O2 , M r 425.92, mp 242.5 ◦ C, avail-


able as maleate (M r 544.06).

Trade names: Gastrozepin (Thomae) and


many others (see [23]).
Trade names: Motilium (Byk Gulden) and
others (see [23]).
3.4.4. Sucralfate
Metoclopramide [364-62-5], 4-ami-
no-5-chloro-N-[2-(diethylamino)-ethyl]-o- Sucralfate [54182-58-0], sucrose hydro-
anisamide (WHO) [155], C14 H22 ClN3 O2 , M r gen sulfate basic aluminum salt (WHO),
299.81, mp 146.5 – 148 ◦ C, solubility in wa- C12 H54 Al16 O75 S8 , M r 2086.74, insoluble in
ter at 25 ◦ C 0.02 wt %, pK a -values 7.3 and water, soluble in dilute HCl or NaOH, pK a
9.0. Metoclopramide is available as hydrochlo- 0.43 – 1.19.
ride monohydrate (M r 354.28) as a 10 % aque-
ous solution of pH 4.5 – 6.0.

Sucralfate was first introduced in Japan in the


late 1960s. Numerous active components of su-
Trade names: Gastrosil (Heumann) and many cralfate have been described which act in con-
others (see [23]). cert to make sucralfate a gastroprotective antiul-
cer drug [159], [160] that even displays “cy-
toprotective” actions (i.e., on a cellular level)
3.4.3. Antimuscarinic Drugs in vitro [161]. Maintenance of mucosal blood
flow ensures rapid repair of mucosal damage;
The only antimuscarinic drug still used in increased mucosal bicarbonate and mucus se-
peptic ulcer disease is pirenzepine which has cretion protects the mucosa; and binding of fi-
limited selectivity for M1 -muscarinic recep- broblast growth factor and epidermal growth
tors present on both intramural neurons and factor to the sucralfate gel adhering to the in-
paracrine, histamine-releasing cells (i.e., ECL- jured mucosa enhances angiogenesis and epithe-
cells) of the gastric mucosa [156], [157]. lization of ulcerated areas [160]. Stimulation of
PG biosynthesis and release of somatostatin may
Pirenzepine [28797-61-7], 5,11-dihydro- contribute to these actions and may further ex-
11-[(4-methyl-1-piperazinyl)acetyl]-6H- plain reduction of both parietal cell responsive-
pyrido[2,3-b][1,4]benzodiazepin-6-one (WHO) ness and ulcer relapse rate following ulcer heal-
[158], C19 H21 N5 O2 , M r 351.42, pK a values 2.1 ing by sucralfate [159]. These effects may also
and 8.1. Pirenzepine is available as dihydrochlo- explain reduced acid secretion under sucralfate
ride monohydrate, M r 442.36, soluble in water. therapy [162]. In addition, sucralfate may inter-
Because of its unfavorable balance between fere with the attachment of H.p. to the gastric
weak antisecretory and frequent anticholiner- epithelium and may thereby suppress to some
gic adverse effects (dry mouth, blurred vision), extent (but not eradicate) H.p. infection [159],
pirenzepine is considered obsolete [128]. [162]. Healing rates with sucralfate are simi-
lar to, and ulcer relapse rates are even lower
than those after, H2 -receptor antagonists [132],
Antiulcer Drugs 13

[163]. Tolerability is excellent, with aluminum- Bismuth subsalicylate [14882-18-9],


induced constipation being the most prominent C7 H5 BiO4 .
side effect seen in 2 – 4 % of the patients [132], Trade names: Jatrox (Procter & Gamble) and
[164]. others (see [23]).
Trade names: Ulcogant (Merck) and many
others (see [23]). Mode of Action. These colloidal, basic bis-
muth preparations are, with the exception of bis-
muth citrate, almost insoluble in water. Depend-
3.4.5. Bismuth Compounds ing on the actual pH, they form either colloidal
solutions, highly hydrated gels, or thin films
A variety of bismuth compounds are available [167]. These cover the ulcerated areas, predom-
as antiulcer drugs. inantly due to formation of bismuth oxychlo-
ride BiOCl, but also more complex structures
Bismuth aluminate [12284-76-3], containing free COOH and BiO+ groups. These
Bi2 (Al2 O4 )3 . structures are precipitated from bismuth salts by
Trade name: Noemin (Trommsdorf) and oth- gastric acid. Bismuth citrate is highly soluble in
ers (see [23]). water but precipitates in an acid medium when
the solubility product of its free acid (formed ac-
Bismuth citrate is on the market as a col- cording to R–CH2 CO− +
2 + H  R–CH2 CO2 H)
loidal ammonium – potassium salt which is a is exceeded [168]. Apart from the tenacious
mixture of the corresponding salts of the bismuth layer formed on the mucosa at low pH (see
citrate complex [BiC6 H4 O7 ]− (K or NH4 )+ above), bismuth salts display a variety of addi-
(see below) and citric acid [C6 H5 O7 ]3− 3(K or tional action components that may be partially
NH4 )+ . The so-called “colloidal bismuth subci- related to their reaction with thiol groups [169]
trate, CBS” in fact contains the aforementioned and proteins [170], [171]. These additional ef-
bismuth citrate complex [165] in which H+ of fects include stimulation of PGE2 biosynthesis
one citric acid carboxyl group is substituted by and bicarbonate secretion, and either stimula-
either K+ or NH+ 4 . These units are further com-
tion of mucus secretion or inhibition of its en-
plexed to a colloidal material [166]. Bismuth cit- zymatic degradation. Bismuth compounds also
rate is highly soluble in water. depress pepsin activity by either chief cell inhi-
bition or pepsin complexation, and inhibit many
enzymes of H.p., thus limiting mucus degra-
dation. Bismuth salts bind and thereby inacti-
vate bile acids at low pH, and accumulate epi-
dermal growth factor in the ulcer area which
may accelerate healing due to its trophic ac-
tions. Bismuth salts exert antimicrobial activity,
Trade names: Telen (Yamanouchi) and others also in H.p. infections [172]. The above phar-
(see [23]). macodynamics of bismuth have been reviewed
in [168], [173], with special reference to bis-
Bismuth subgallate [99-26-3], C7 H7 BiO7 , muth citrate, and in general in [132]. Substantial
lipid solubility leads to a high rate of absorption, bismuth concentrations in the mucus or within
in contrast to the other salts (but see bismuth cit- the mucosa are observed for only a few hours
rate). following administration. Therefore, the low ul-
Trade names: Bismofalk (Falk) and others cer relapse rate with bismuth compounds may
(see [23]). partially be due to trace amounts of bismuth
systematically absorbed and, due to slow ex-
Bismuth subnitrate [1304-85-4], cretion, still available in the gastric tissue over
BiO(NO3 )·H2 O. up to 3 months. Nonetheless, safety of therapy
Trade name: Angass (Medice). with bismuth compounds seems to be excellent
with only mild and infrequent side effects [174].
Probably due to its pK a value in the order of 3
14 Antiulcer Drugs

and, therefore, its comparatively fair ability to of the total pumps in a parietal cell during meal
penetrate the stomach wall under acidic condi- stimulation [33], [177]. APAs will therefore re-
tions as an uncharged molecule, bismuth citrate sult in a more rapid and complete inhibition of
may produce higher (although transient) peak acid secretion than PPIs on a first administration.
plasma concentrations of bismuth than some of The duration of this inhibition by APAs corre-
the other bismuth salts which are unstable un- sponds to the drug’s actual plasma concentration
der these conditions. However, no major toxic as opposed to a prolonged inhibition with cova-
effects have been reported [174]. lently binding PPIs [178], [179]. APAs with dif-
ferent chemical structures have been synthesized
Ranitidine bismuth citrate (RBC) [128345- (for example, imidazopyridines and acylquino-
62-0], C19 H27 BiN4 O10 S, M r 712.48, is a novel lines; see [178]) but not one has reached the mar-
salt of the base ranitidine and the monobasic ket so far. Although these weak nitrogen bases,
acid bismuth citrate (note that a widely used like the PPIs, accumulate within the acidic space
nomenclature is subcitrate instead of citrate, see of the parietal cell, their selectivity for the acid
above and [165]). Ranitidine bismuth citrate pump is based on a specific fit between their
forms a suspension in water but is soluble at chemical structure and that of the acid pump,
low pH values to release its components. Ac- being in line with a competitive mode of action.
cording to [175] RBC inactivates pepsin and
H.p. more efficiently than an admixture of its Pumaprazole [158364-59-1], methyl
components due to its better solubility. How- 2-[[(2,3-dimethylimidazo[1,2-a]pyridin-8-
ever, as discussed in page 13, bismuth citrate yl)amino]methyl]-3-methylcarbanilate (WHO),
released at low pH will again precipitate as C19 H22 N4 O2 . Pumaprazole has been aban-
soon as the solubility product of its free acid is doned in favor of a pharmacologically better
exceeded. Ranitidine bismuth citrate has been follow up compound presently under develop-
developed for combination therapy with antibi- ment at Byk Gulden. The WHO decided to
otics for eradication of H.p. Depending on the label the APAs by the stem syllable “-prazole”
co-prescription, eradication ranged from 15 to in order “to maintain the number of stems at
92 % in an intention-to-treat analysis [176]. It is a manageable level”, irrespective of the com-
claimed to combine the antisecretory action of pletely different (i.e., reversible vs. covalent)
ranitidine with the antimicrobial and gastropro- modes of drug – target interactions by APAs and
tective actions of bismuth. PPIs (WHO decision communicated on Nov. 30,
Trade name: Pylorid (GlaxoWellcome). 1995). Unfortunately, this pharmacologically
misleading but legally binding decision will con-
fuse the rational use and differential perception
4. Prospect for Future Developments of APAs vs. PPIs in the future and counteracts
the effort for an unambiguous terminology in
pharmacology. It should be noted that the terms
4.1. Reversibly Binding Acid Pump APAs and PPIs have been coined specifically
Antagonists to make a clearly recognizable distinction be-
tween the two chemically and mechanistically
In contrast to PPIs which bind covalently to the different classes of drugs.
proton (acid) pump (see Section 3.1), acid pump
antagonists (APAs) bind reversibly and compete
with potassium on the luminal (extracytoplas- 4.2. Drugs Directed Against
mic) side of the H+ /K+ -ATPase to interrupt its Helicobacter pylori
catalytic cycle [20]. This reversible blockade not
only regards the activated pumps already incor- Helicobacter pylori (H.p.) has been recognized
porated into the apical membrane, but also the as a causal factor of most peptic ulcers (see
resting pumps [177] still present in cytoplasmic Chap. 2). While companies worldwide try to
vesicles. The latter are devoid of any proton gra- develop antimicrobials both specific for H.p.
dient, they are unable to activate PPIs for this and effective as a monotherapy, current ther-
reason, and probably amount to about one-third apy still takes advantage of the available reper-
Antiulcer Drugs 15

toire of antibiotics [180–183] (→ Antibiotics). onists [196]. Of course, also PPIs are very ef-
Although it has been claimed that antibiotics fective prophylactic drugs [197]. Antacids have
alone can eradicate H.p. infection and heal DU maintained their place in “common heartburn”,
[184], present eradication regimes include one gastritis, and less severe cases of GERD or GU
of the PPIs (see [180–183]) to enhance the effect and DU [132], although they constitute an in-
of the antibiotics by a mode of interaction still ferior treatment option. Their major application
under debate [185]. One-week triple therapies will only be as cheap self-medication remedies.
consisting of a PPI in combination with two of
the three antibiotics amoxicillin (→ Antibiotics,
Chap. 4.), clarithromycin, and metronidazole 6. References
(→ Imidazole and Derivatives, Chap. 4.3.) are
the therapies of choice according to the Maas- 1. P. J. Kahrilas: “Gastroesophageal reflux
tricht consensus report [186]. disease,” in G. Friedman, E. D. Jacobson,
R. W. McCallum (eds.): Gastrointestinal
Pharmacology and Therapeutics,
5. Relative Drug Assessment Lippincott-Raven Publishers, Philadelphia
1997, pp. 31 – 44.
Burget et al. [187] have published a model 2. P. Malfertheiner, J. W. Freston: “Helicobacter
predicting the healing rates in DU, increasing pylori-induced gastritis, ulceration and
up to an optimum defined by both a pH thresh- neoplasia,” in G. Friedman, E. D. Jacobson,
old and the percentage time the intragastric pH R. W. McCallum (eds.): Gastrointestinal
was above this threshold during a 24-h-period. A Pharmacology and Therapeutics,
similar relationship in GERD treatment was later Lippincott-Raven Publishers, Philadelphia
1997, pp. 21 – 29.
reviewed by Hunt [188]. It generally means that
3. C. W. Howden: “Gastric and Duodenal ulcers,”
the longer the duration and the higher the degree
in G. Friedmann, E. D. Jacobson, R. W.
of pH elevation by a certain drug is, the better McCallum (eds.): Gastrointestinal
the healing rate will be, up to an optimum which Pharmacology and Therapeutics,
depends on the particular disease (GERD, GU, Lippincott-Raven Publishers, Philadelphia
DU, Zollinger – Ellison Syndrome). This clearly 1997, pp. 45 – 51.
favors the PPIs (see Section 3.1) for treatment 4. N. M. Agrawal, E. Z. Dajani: “Drug-induced
of the H.p.-unrelated reflux esophagitis [189], Ulcerogenesis,” in G. Friedman, E. D.
[190]. The superiority of PPIs also shows in Jacobson, R. W. McCallum (eds.):
treatment of GU and DU with the limitation that Gastrointestinal Pharmacology and
any therapy eradicating H.p. (see Section 4.2) or Therapeutics, Lippincott-Raven Publishers,
suppressing H.p. in addition to gastroprotective Philadelphia 1997, pp. 55 – 63.
effects (see Sections 3.4.4 and 3.4.5) seems to 5. P. N. Maton: “Management of acid
reduce the ulcer relapse rate compared to exclu- hypersecretion in patients with
sively antisecretory drugs [191]. An interesting Zollinger-Ellison syndrome,” in G. Friedman,
observation in this context is that eradication of E. D. Jacobson, R. W. McCallum (eds.):
H.p. may provoke GERD [192] or DU [193]. Gastrointestinal Pharmacology and
This may be due to the loss of neutralizing (NH3 ) Therapeutics, Lippincott-Raven Publishers,
or antisecretory compounds that had been pro- Philadelphia 1997, pp. 65 – 73.
6. D. J. Cook: “Stress Ulcer Prophylaxis,” in G.
duced by H.p. in the patient’s stomach before
Friedman, E. D. Jacobson, R. W. McCallum
eradication therapy [194]. Obviously, this ob- (eds.): Gastrointestinal Pharmacology and
servation of DU induction by H.p. eradication Therapeutics, Lippincott-Raven Publishers,
therapy relates to the well known, low percent- Philadelphia 1997, pp. 75 – 78.
age of peptic ulcer cases causally independent 7. J. H. Kurata, Gastroenterology 96 (1989)
of H.p. infection. 569 – 580.
NSAID-induced peptic ulcers [195] can be 8. K. H. Holtermüller: “Magen-Darm-Mittel,” in
prevented by prophylactic treatment with sub- U. Schwabe, D. Paffrath (eds.):
antisecretory doses of misoprostol (see Sec- Arzneiverordnungs-Report ’96, Gustav
tion 3.4.1) but less so with H2 -receptor antag- Fischer Verlag, Stuttgart 1996, pp. 305 – 324.
16 Antiulcer Drugs

9. J. H. Kurata, A. N. Nogawa, J. Clin. 30. K. Gedda, D. Scott, M. Besancon, G. Sachs,


Gastroenterol. 24 (1997) 2 – 17. Gastroenterology 106 (1994) Suppl., A79.
10. N. Banatvala, K. Mayo, F. Mégraud, R. 31. K. Gedda et al., Gastroenterology 109 (1995)
Jennings, J. J. Deeks, R. A. Feldmann, J. 1134 – 1141.
Infect. Dis. 168 (1993) 219 – 221. 32. H. Li, H. F. Helander, Scand. J. Gastroenterol.
11. D. J. E. Cullen, B. J. Collins, K. J. 30 (1995) 205 – 209.
Christiansen, J. Epis, J. R. Warren, I. Surveyor, 33. B. I. Hirschowitz et al., Dig. Dis. Sci. 40
K. J. Cullen, Gut 34 (1993) 1681 – 1682. (1995) Suppl 2, 3S – 23S.
12. P. Sahay, A. T. R. Axon, Helicobacter 1 34. S. Postius, U. Bräuer, W. Kromer, Life Sci. 49
(1996) 175 – 182. (1991) 1047 – 1052.
13. F. Hornemann, M. Nilius, P. Malfertheiner, P. 35. L. B. Barradell, D. Faulds, D. McTavish,
Bartmann, Helicobacter 2 (1997) 176 – 179. Drugs 44 (1992) 225 – 250.
14. K. Schwarz, Beitr. Klin. Chir. 67 (1910) 36. S. Yasuda et al., Int. J. Clin. Pharmacol. Ther.
96 – 128. 32 (1994) 466 – 473.
15. A. H. Soll: “Gastric duodenal and stress ulcer,” 37. W. Kromer, U. Krüger, R. Huber, M.
in M. H. Sleisenger, J. S. Fordtran (eds.): Hartmann, V. W. Steinijans, Pharmacology 56
Gastrointestinal Disease, 5th ed., vol. 1, W. B. (1998) 57 – 70.
Saunders Company, Philadelphia 1993, 38. M. Hell, K. Eggert, G. Brunner, F. W. Schmidt,
pp. 580 – 679. Z. Gastroenterol. 9 (1989) 562.
16. J. L. Wallace, C. J. Bell, Current opinion in 39. H. G. Dammann et al., Aliment. Pharmacol.
Gastroenterology 12 (1996) 503 – 511. Ther. 7 (1993) 191 – 196.
17. S. J. Konturek, Scand. J. Gastroenterol. 25 40. H. Koop et al., Gut 35 (1994) Suppl 4, A79.
(1990) Suppl 174, 15 – 28. 41. H. Koop et al., Eur. J. Gastroenterol. Hepatol.
18. M. Guslandi, Drugs 41 (1991) 507 – 513. 8 (1996) 915 – 918.
19. K. E. L. McColl, G. M. Fullarton, Gut 34 42. H. E. Scholtz, B. H. Meyer, H. G. Luus, S. Afr.
(1993) 1300 – 1302. Med. J. 85 (1995) 915.
20. G. Sachs, J. M. Shin, C. Briving, B. Wallmark, 43. M. Hartmann et al., Aliment. Pharmacol. Ther.
S. Hersey, Annu. Rev. Pharmacol. Toxicol. 35 10 (1996) 359 – 366.
(1995) 277 – 305. 44. G. Brunner, H. Danz-Neef, C. Athmann, N.
21. L. L. Burnton: “Agents for control of gastric Samayoa, Can. J. Gastroenterol. 11 (1997)
acidity and treatment of peptic ulcers,” in J. G. Suppl A, S2.
Hardman, L. E. Limbird, P. B. Molinoff, R. W. 45. D. J. Hetzel et al., Gastroenterology 95 (1988)
Ruddon, A. G. Goodman (eds.): Goodman & 903 – 912.
Gilman’s The Pharmacological Basis of 46. L. S. Laursen et al., Gastroenterology 102 (II)
Therapeutics, 9th ed., McGraw-Hill, New (1992) Suppl., A110.
York 1996, pp. 901 – 915. 47. C. M. Bate et al., Aliment. Pharmacol. Ther. 7
22. E. Haen, W. Forth: Wirkstoffprofile der (1993) 501 – 507.
Arzneimitteltherapie, Ecomed 48. K. Lauritsen et al., Eur. J. Gastroenterol.
Verlagsgesellschaft, Aulendorf 1997. Hepatol. 4 (1992) 995 – 1000.
23. ABDATA Pharma-Daten-Service: “List of 49. A. Walan et al., N. Engl. J. Med. 320 (1989)
Pharmaceutical Substances,” 69 – 75.
Eschborn/Taunus, Germany, 1997. 50. J. E. Valenzuela et al., Am. J. Gastroenterol.
24. J. E. F. Reynolds (ed.): Martindale. The Extra 91 (1996) 2516 – 2522.
Pharmacopoeia, 31st ed., Royal 51. L. Witzel, H. Gütz, W. Hüttermann, W.
Pharmaceutical Society, London 1996. Schepps, Aliment. Pharmacol. Ther. 9 (1995)
25. Merck Index, 12th edition, Merck & Co., Inc., 19 – 24.
Whitehouse Station N.J. 1996. 52. M. I. Wilde, D. McTavish, Drugs 48 (1994)
26. Index Nominum International Drug Directory, 91 – 132.
16th ed., Medpharm Scientific Publishers, 53. A. Fitton, L. Wiseman, Drugs 51 (1995)
Stuttgart 1995. 460 – 482.
27. W. Kromer, Digestion 56 (1995) 443 – 454. 54. H.-G. Dammann, J. Pharmacol. Ther. 4
28. V. Figala et al., J. Chem. Soc. Chem. Commun. (1995) 114 – 116.
(1986) 125 – 127. 55. P. Müller, B. Simon, Münch. med. Wschr. 138
29. P. Lindberg et al., J. Med. Chem. 29 (1986)
(1996) 802 – 804.
1327 – 1329.
Antiulcer Drugs 17

56. W. Kromer et al., J. Pharmacol. Exp. Ther. 81. S. Joelson et al., Digestion 51 (1992) Suppl 1,
254 (1990) 129 – 135. 93 – 101.
57. U. A. Meyer, Yale J. Biol. Med. 69 (1996) 82. R. Arnold, Aliment. Pharmacol. Ther. 8
203 – 209. (1994) Suppl 1, 65 – 70.
58. W. A. Simon, Naunyn-Schmiedebergs Arch. 83. N. D. Yeomans, Adverse Drug React. Toxicol.
Pharmacol. 353 (1996) Suppl, R10. Rev. 13 (1994) 145 – 156.
59. V. W. Steinijans et al., Int. J. Clin. Pharmacol. 84. L. Ekman et al., Scand. J. Gastroenterol. 20
Ther. 34 (1996) S 31 – S 50. (1985) Suppl 108, 53 – 69.
60. H. J. Koch et al., Int. J. Clin. Pharmacol. Ther. 85. B. K. Sharma et al., Br. Med. J. 289 (1984)
34 (1996) 420 – 423. 717 – 719.
61. R. Gugler, J. C. Jensen, Gastroenterology 89 86. H. Koop, Aliment. Pharmacol. Ther. 6 (1992)
(1985) 1235 – 1241. 399 – 406.
62. R. Gugler, J. C. Jensen, Pharmacol. Ther. 33 87. D. Diaz et al., Gastroenterology 99 (1990)
(1987) 133 – 137. 737 – 747.
63. T. Andersson, C. Cederberg, G. Edvardsson, 88. W. M. McDonnell, J. M. Scheiman, P. G.
A. Heggelund, P. Lundborg, Clin. Pharmacol. Traber, Gastroenterology 103 (1992)
Ther. 47 (1990) 79 – 85. 1509 – 1516.
64. T. Andersson et al., Eur. J. Clin. Pharmacol. 89. B. Burlinson, S. H. Morriss, D. G. Gatehouse,
39 (1990) 51 – 54. D. J. Tweats, Lancet 335 (1990) 419.
65. Y. Caraco, T. Tateishi, A. J. J. Wood, Clin. 90. W. Creutzfeldt, Drug Saf. 10 (1994) 66 – 82.
Pharmacol. Ther. 58 (1995) 62 – 72. 91. W. Creutzfeldt, Z. Gastrenterol. 33 (1995)
66. M. U. R. Naidu et al., Drug Invest. 7 (1994) Suppl 1, 12 – 21.
8 – 12. 92. W. C. Creutzfeldt, A. Blum, Lancet 343
67. P. J. Prichard et al., Br. J. Clin. Pharmacol. 24 (1994) 1098.
(1987) 543 – 545. 93. D. Colin-Jones, Lancet 343 (1994) 1369.
68. S. Toon, B. L. Holt, F. G. P. Mullins, A. Khan, 94. S. Holt, Zhu Zhao-Hua, R. E. Powers,
J. Pharm. Pharmacol. 47 (1995) 85 – 88. Gastroenterology 101 (1991) 650 – 656.
69. T. Sutfin et al., Ther. Drug Monit. 11 (1989) 95. W. Schepp, V. Schusdziarra, Z. Gastroenterol.
176 – 184. 28 (1990) 644 – 645.
70. P. Unge et al., Br. J. Clin. Pharmacol. 34 96. J. Fryklund, A.-K. Falknäs, H. F. Helander,
(1992) 509 – 512. Scand. J. Gastroenterol. 27 (1992) 521 – 528.
71. K. Zech et al., J. Clin. Pharmacol. Ther. 34 97. F. J. Garrote et al., Lancet 340 (1992) 672.
(1996) Suppl 1, S3 – S6. 98. F. Sarrot-Reynauld, H. Charras-De Matteis, C.
72. B. Simon et al., Aliment. Pharmacol. Ther. 4 Massot, Reactions 648 (1997) 10.
(1990) 373 – 379. 99. R. M. Ricci, K. C. Deering, Cutis 57 (1996)
73. H. Bliesath, R. Huber, M. Hartmann, R. 434.
Lühmann, W. Wurst, Int. J. Clin. Pharmacol. 100. M. Beutler, K. Hartmann, M. Kuhn, J.
Ther. 32 (1994) 44 – 50. Gartmann, Br. Med. J. 309 (1994) 1620.
74. R. Huber et al., Int. J. Clin. Pharmacol. Ther. 101. C. Buckley, Br. J. Dermatol. 135 (1996)
34 (1996) 185 – 194. 660 – 661.
75. T. Andersson, C. Cederberg, A. Heggelund, P. 102. H. Amjad, W. Beckley, Blood 86 (1995)
Lundborg, Drug Invest. 3 (1991) 45 – 51. Suppl 1, 846A.
76. T. Andersson, C. Cederberg, C.-G. Regårdh, I. 103. S. J. Ruffenach, M. S. Siskind, Y. H. H. Lien,
Skåndberg, Eur. J. Clin. Pharmacol. 39 Am. J. Med. 93 (1992) 472 – 473.
(1990) 195 – 197. 104. M. Assouad, S. L. Vicks, M. V. Pokroy, P. J.
77. A. E. Zimmermann, B. G. Katona, Willcourt, Lancet 344 (1994) 549.
Pharmacotherapy 17 (1997) 308 – 326. 105. E. J. M. Meeuwisse et al., Reactions 639
78. H. Nagaya, N. Inatomi, A. Nohara, H. Satoh, (1997) 10.
Biochem. Pharmacol. 42 (1991) 1875 – 1878. 106. J. M. Prignet, E. Chauveau, J. L. Duval, D.
79. S. Allenmark, B. Bomgren, H. Boren, P.-O. Kunkel, Semain des Hospitaux 72 (1996)
Lagerström, Anal. Biochem. 136 (1984) 19 – 20.
293 – 297. 107. A. Kraus, L. F. Flores Suarez, Lancet 345
80. P. Erlandsson, R. Isaksson, P. Lorentzon, P. (1995) 461 – 462.
Lindberg, J. Chromatogr. 532 (1990) 108. W. Kromer, B. Kohl, Drugs Future 15 (1990)
305 – 319. 801 – 804.
18 Antiulcer Drugs

109. W. Kromer, S. Gönne, R. Riedel, S. Postius, 135. U. Assmus, A. Schwind, Pharma


Pharmacology 41 (1990) 333 – 337. International 5 (1990) 172 – 178.
110. R. Huber et al., Aliment. Pharmacol. Ther. 9 136. J. Monés et al., Gut 32 (1991) 147 – 150.
(1995) 363 – 378. 137. G. Brown, M. C. Gastuche, Clay Miner. 7
111. V. W. Steinijans (ed.), Int. J. Pharmacol. Ther. (1967) 193 – 201.
34 (1996) Suppl 1. 138. H. F. W. Taylor, Mineral. Mag. 39 (1973)
112. J. W. Black, W. A. M. Duncan, C. J. Durant, 377 – 389.
C. R. Ganellin, E. M. Parsons, Nature 236 139. L. R. Gutermann, C. J. Falzone, G. E. Wilson,
(1972) 385 – 390. J. Pharm. Sci. 75 (1986) 502 – 505.
113. K.-Fr. Sewing, Scand. J. Gastroenterol. 23 140. H. C. Dollinger, E. Holzberg: “Absorption von
(1988) Suppl 146, 73 – 77. Aluminium und Magnesium unter Magaldrat
114. J. G. Mills et al., Aliment. Pharmacol. Ther. und anderen magnesium-aluminiumhaltigen
11 (1997) 129 – 137. Antazida,” in R. Arnold, W. H. Häcki (eds.):
115. G. Bianchi Porro, F. Parente, Scand. J. Magaldrat, Verlag Christian M. Silinsky,
Gastroenterol. 23 (1988) Suppl 153, 81 – 88. Nürnberg 1985, pp. 98 – 103.
116. C. U. Nwoloko, J. T. L. Smith, A. Sawyerr, 141. B. Winterberg et al., J. Trace Elem.
R. E. Pounder, Aliment. Pharmacol. Ther. 4 Electrolytes Health Dis. 1 (1987) 69 – 72.
(1990) Suppl 1, 29 – 46. 142. W. H. Häcki, F. Conti, M. Uehliger:
117. C. H. Wilder-Smith, H. S. Merki, Scand. J. “Diskrepanz zwischen In-vivo- und
Gastroenterol. 27 (1992) Suppl 193, 14 – 19. In-vitro-Säurebindungskapazitäten von
118. M. Murray, Drug Metab. Rev. 18 (1987) Magaldrat (Riopan), Gelusil und Maaloxan,”
55 – 81. in R. Arnold, W. H. Häcki (eds.): Magaldrat,
119. R. N. Brogden, R. C. Heel, T. M. Speight, G. S. Verlag Christian M. Silinsky, Nürnberg 1985,
Avery, Drugs 15 (1978) 93 – 131. pp. 57 – 61.
120. M. Freemantle, C&E News (1998) Jan. 5, 143. C. Baur, A. Becker, R. Linder, T. Schwan,
31 – 32. Drug Res. 31 (1981) 504 – 507.
121. D. M. Campoli-Richards, S. P. Clissold, Drugs 144. J. L. Wallace, A. W. Tigley, Aliment.
32 (1986) 197 – 221. Pharmacol. Ther. 9 (1995) 227 – 235.
122. A. H. Price, R. N. Brogden, Drugs 36 (1988) 145. S. J. Sontag, Drugs 32 (1986) 445 – 457.
521 – 539. 146. K. L. Goa, J. P. Monk, Drugs 34 (1987)
123. S. M. Grant, H. D. Langtry, R. N. Brogden, 539 – 559.
Drugs 37 (1989) 801 – 870. 147. J. P. Monk, S. P. Clissold, Drugs 33 (1987)
124. D. Murdoch, D. McTavish, Drugs 42 (1991) 1 – 30.
240 – 260. 148. B. Ramirez, J. E. Richter, Aliment. Pharmacol.
125. C.-K. Ching, S.-K. Lam, Drugs 47 (1994) Ther. 7 (1993) 5 – 20.
305 – 317. 149. M. R. Briejer, L. M. A. Akkermans, J. A. J.
126. R. P. Walt, M. J. S. Langman, Drugs 42 (1991) Schuurkes, Pharmacol. Res. 47 (1995)
205 – 212. 631 – 651.
127. K. D. Fine, G. J. Krejs, J. S. Fordtran: 150. K.-A. Gwee, N. W. Read, Aliment. Pharmacol.
“Diarrhea,” in M. H. Sleisenger, J. S. Fordtran Ther. 8 (1994) 105 – 118.
(eds.): Gastrointestinal Disease, W. B. 151. F. Mitchelson, Drugs 43 (1992) 295 – 315.
Saunders, Philadelphia 1993, pp. 1043 – 1072. 152. L. R. Wiseman, D. Faulds, Drugs 47 (1994)
128. D. W. Piper, Drug Saf. 12 (1995) 120 – 138. 116 – 152.
129. W. Forth, Dt. Ärztebl. 84 (1987) 153. J. M. C. Greif, D. Rowbotham, Clin.
C-1316 – 1317. Pharmacokinet. 27 (1994) 447 – 461.
130. I. Szelenyi, Drugs News Perspect. 3 (1990) 154. M. C. Champion, Pharma 19 (1988)
396 – 402. 499 – 505.
131. H. Arnott, On Contin. Pract. 18 (1991) 155. R. A. Harrington et al., Drugs 25 (1983)
36 – 44. 451 – 494.
132. Van Ness et al. (eds.): Handbook of 156. E. C. Hulme, N. J. M. Birdsall, N. J. Buckley,
Gastrointestinal Drug Therapy, 2nd ed., Annu. Rev. Pharmacol. Toxicol. 30 (1990)
Little, Brown and Company, Boston 1995. 633 – 673.
133. W. Kurtz, Fortsch. Med. 111 (1993) 93 – 96. 157. W. Kromer, E. Baron, R. Boer, M. Eltze,
134. U. Krüger, Gastro-Entero-Hepatologie 3 Naunyn-Schmiedebergs Arch. Pharmacol. 343
(1985) Suppl 1, 5 – 7. (1991) 7 – 13.
Antiulcer Drugs 19

158. A. A. Carmine, R. N. Brogden, Drugs 30 178. A. J. Pope, M. E. Parsons, Trends Pharmacol.


(1985) 85 – 126. Sci. 14 (1993) 323 – 325.
159. W. D. W. Rees, Scand. J. Gastroenterol. 27 179. W. Wurst, M. Hartmann, Yale J. Biol. Med. 69
(1992) Suppl 191, 4 – 6. (1996) 233 – 243.
160. S. Szabo, Scand. J. Gastroenterol 26 (1991) 180. A. T. R. Axon, Scand. J. Gastroenterol. 29
Suppl 185, 7 – 12. (1994) Suppl 201, 16 – 23.
161. M. Romano, M. Razandi, K. J. Ivey, Dig. Dis. 181. A. Hackelsberger, P. Malfertheiner, Drug Saf.
Sci. 35 (1990) 467 – 476. 15 (1996) 30 – 52.
162. S. Banerjee et al., Gastroenterology 110 182. J. G. Pentson, Aliment. Pharmacol. Ther. 10
(1996) 717 – 724. (1996) 469 – 486.
163. T. E. Bynum, Scand. J. Gastroenterol. 27 183. F. Mégraud, Aliment. Pharmacol. Ther. 11
(1992) Suppl 191, 1 – 3. (1997) Suppl 1, 43 – 53.
164. I. N. Marks, Scand. J. Gastroenterol. 26 184. S.-K. Lam et al., Gut 41 (1997) 43 – 48.
(1991) Suppl 185, 36 – 42. 185. W. L. Peterson, Aliment. Pharmacol. Ther. 11
165. W. A. Herrmann, E. Herdtweck, L. Pajdla, (1997) Suppl 1, 21 – 25.
Inorg. Chem. 30 (1991) 2579 – 2581. 186. EHPSG (The European Helicobacter Pylori
166. E. Asato, K. Katsura, M. Mikuriya, T. Fujii, J.
Study Group) Gut 41 (1997) 8 – 13.
Reedijk, Inorg. Chem. 32 (1993) 5322 – 5329.
187. D. W. Burget, S. G. Chiverton, R. H. Hunt,
167. R. Raedsch, Dtsch. med. Wschr. 116 (1991)
Gastroenterology 99 (1990) 345 – 351.
821 – 824.
168. A. J. Wagstaff, P. Benfield, J. P. Monk, Drugs 188. R. H. Hunt, Aliment. Pharmacol. Ther. 9
36 (1988) 132 – 157. (1995) Suppl 1, 3 – 7.
169. W. Beil, S. Bierbaum, K.-F. Sewing, 189. E. C. Klinkenberg-Knol, H. P. M. Festen,
Pharmacology 47 (1993) 135 – 140. S. G. M. Meuwissen, Drugs 49 (1995)
170. W. Beil, S. Bierbaum, K.-F. Sewing, 695 – 710.
Pharmacology 47 (1993) 141 – 144. 190. N. Chiba, Ch. J. De Gara, J. M. Wilkinson,
171. J. Wieriks et al., Scand. J. Gastroenterol. 17 R. H. Hunt, Gastroenterology 112 (1997)
(1982) Suppl 80, 11 – 16. 1798 – 1810.
172. J. R. Lambert, P. Midolo, Aliment. Pharmacol. 191. M. G. Korman, Scand. J. Gastroenterol. 30
Ther. 11 (1997) Suppl 1, 27 – 33. (1995) Suppl. 208, 21 – 23.
173. S. P. Lee, Scand. J. Gastroenterol. 26 (1991) 192. J. Labenz et al., Gastroenterology 112 (1997)
Suppl 185, 1 – 6. 1442 – 1447.
174. L. A. Tillman, F. M. Drake, J. S. Dixon, J. R. 193. T. Pohle, E. Hilker, W. Domschke, Z.
Wood, Aliment. Pharmacol. Ther. 10 (1996) Gastroenterol. 35 (1997) 563 – 565.
459 – 467. 194. E. F. Verdú et al., Gut 37 (1995) 743 – 748.
175. A. A. McColm et al., Aliment. Pharmacol. 195. J. M. Scheiman, Dig. Dis. 12 (1994) 210 – 222.
Ther. 10 (1996) 241 – 250. 196. M. Koch et al., Dig. Dis. 13 (1995) Suppl 1,
176. J. W. Wyeth et al., Aliment. Pharmacol. Ther. 62 – 74.
10 (1996) 623 – 630. 197. A. Garner, H. Fadlallah, Exp. Opin. Invest.
177. K. B. Munson, G. Sachs, Biochemistry 27 Drugs 6 (1997) 885 – 893.
(1988) 3932 – 3938.

Aldols → Aldehydes, Aliphatic and Araliphatic


Alfol Process → Fatty Alcohols
Algin → Polysaccharides
Alkali and Chlorine Products → Chlorine
Alkali and Chlorine Products → Sodium Hydroxide

You might also like