throbber
GASTROENTEROLOGY 2000;118:S9–S31
`
`Acid Suppression: Optimizing Therapy for Gastroduodenal
`Ulcer Healing, Gastroesophageal Reflux Disease, and
`Stress-Related Erosive Syndrome
`
`M. MICHAEL WOLFE* and GEORGE SACHS ‡
`*Section of Gastroenterology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts; and ‡Division of
`Gastroenterology, UCLA School of Medicine and West Los Angeles VA Medical Center, Los Angeles, California
`
`One of the hallmarks of the mammalian stomach is its
`
`ability to secrete large quantities of concentrated
`(0.16 mol/L) hydrochloric acid (HCl).1 Although it is
`generally assumed that gastric acid and the proteolytic
`enzyme pepsin are required to initiate digestion, achlor-
`hydric individuals generally do not develop malabsorp-
`tion unless small bowel bacterial overgrowth is present. It
`is thus likely that the ability of the stomach to secrete
`acid evolved primarily from a need to sustain a sterile
`intragastric milieu. Organisms that possessed the capac-
`ity to kill ingested bacteria and other microbes were able
`to avoid the development of enteric colonization, and
`thereby ensure both efficient absorption of nutrients and
`prevention of systemic infections.1 Nevertheless, when
`present, gastric acid does play a significant role in protein
`hydrolysis and other aspects of the digestive process, and
`under various conditions, acid may play an etiologic role
`in producing various forms of discomfort and inciting
`esophageal and gastroduodenal mucosal injury.
`The normal human stomach contains approximately 1
`billion parietal cells that secrete hydrogen ions into the
`gastric lumen in response to various physiological stimuli.
`The generation of H1 ions is mediated by 3 pathways:
`neurocrine, paracrine, and endocrine (Figure 1). The
`principal neurocrine transmitter is acetylcholine, which
`is released by vagal postganglionic neurons and appears to
`stimulate H1 ion generation directly via a parietal cell
`muscarinic M3 receptor. Histamine is the primary para-
`crine transmitter that binds to H2-specific receptors on
`parietal cells. Adenylate cyclase is then activated, leading
`to an increase in adenosine 38,58-cyclic monophosphate
`(cAMP) levels and subsequent generation of H1 ions. The
`secretion of gastrin from antral G cells comprises the
`endocrine pathway and stimulates H1 ion generation
`both directly and indirectly, the latter by stimulating
`histamine secretion from enterochromaffin-like (ECL)
`cells of the corpus and fundus. Interactions among
`neurocrine, paracrine, and endocrine pathways are coordi-
`nated to promote or inhibit H1 ion generation. Hista-
`
`mine appears to represent the dominant route, because
`gastrin stimulates acid secretion principally by promot-
`ing histamine release from ECL cells.2,3 Thus, ECL cells
`are often referred to as ‘‘controller’’ cells in the process of
`gastric acid secretion.
`A negative feedback loop governs both gastrin release
`and the return of acid secretion to basal level.1,4–6 This
`autoregulatory mechanism prevents postprandial acid
`hypersecretion. After ingestion of a meal, gastrin release
`stimulates secretion of gastric acid. The intraluminal pH
`begins to decrease, which stimulates release of somatosta-
`tin from antral D cells, possibly through the activation of
`calcitonin gene–related peptide (CGRP) neurons.5,7 So-
`matostatin then appears to act via a paracrine mechanism
`to inhibit further release of gastrin from G cells.8
`Somatostatin produced by D cells in the gastric corpus
`and fundus may also directly inhibit acid secretion from
`parietal cells and may suppress histamine release from
`ECL cells (Figure 1).6,9 Other recent observations indicate
`that several other neurotransmitters, including vasoactive
`intestinal peptide (VIP), galanin, and pituitary adenylate
`cyclase–activating peptide, may play important roles in
`regulating gastric acid secretion, both directly and
`indirectly, under physiological conditions.10
`Pathophysiology of Acid-Related
`Disorders
`Gastroduodenal (Peptic) Ulcer
`The treatment of duodenal ulcer (DU) has served
`as the basis (correctly or incorrectly) for the management
`of nearly all acid-related disorders. This supposition in all
`likelihood contributed to delays in the optimal manage-
`ment of other gastrointestinal (GI) disorders in which
`
`Abbreviations used in this paper: DU, duodenal ulcer; ECL, entero-
`chromaffin-like; GERD, gastroesophageal reflux disease; GI, gastro-
`intestinal; GU, gastric ulcer; NCCP, noncardiac chest pain; PPI,
`proton pump inhibitor; PUD, peptic ulcer disease.
`r 2000 by the American Gastroenterological Association
`0016-5085/00/$10.00
`
`DRL EXHIBIT 1014 PAGE 1
`
`

`

`S10 WOLFE AND SACHS
`
`GASTROENTEROLOGY Vol. 118, No. 2
`
`Figure 1. Schematic representa-
`tion of the factors influencing
`gastric acid secretion by the pa-
`rietal cell, depicting neurocrine
`(acetylcholine and other neuro-
`transmitters from vagal efferent
`neurons), paracrine (somatosta-
`tin from D cells and histamine
`from gastric ECL cells), and en-
`docrine (circulating gastrin) fac-
`tors. Dashedarrowsindicate po-
`tential sites of pharmacological
`inhibition of acid secretion, ei-
`ther via receptor antagonism or
`via inhibition of H1,K1-ATPase.
`A, acetylcholine and other neuro-
`transmitters; H, histamine; G,
`gastrin; L365,260, synthetic
`gastrin receptor antagonist; PG,
`prostaglandin; S, somatostatin.
`
`cells, which decreases the magnitude of the response to
`luminal acidification.12–14 Thus,
`in patients with H.
`infection limited to the antrum, the negative
`pylori
`feedback inhibition of gastrin release is attenuated,
`resulting in higher postprandial gastrin levels and hyper-
`secretion of acid.
`Despite the existence of meal-induced hyperchlorhy-
`dria in DU patients, the presence of food in the stomach
`has a buffering effect that may protect the gastroduodenal
`mucosa from acid-induced injury. However, at night and
`during other prolonged periods of fasting, acid bathes the
`‘‘bare’’ mucosa, and in DU patients, the increase in
`nocturnal acid secretion magnifies this effect. Duodenal
`bicarbonate secretion also appears to be impaired in
`patients with DU,15 as well as in those infected with
`H. pylori, making the mucosal exposure to acid even
`greater. These observations, as discussed below, form the
`rationale for single nocturnal dosing of H2-receptor
`antagonists in the treatment of DU, a mode of therapy
`that is at least as effective as multiple dosing regimens.
`Clearly, factors other than acid and pepsin are involved
`in the pathogenesis of peptic ulcer disease (PUD), because
`only 30% of patients with DUs and very few patients
`with GUs are hyperchlorhydric.1 The balance between
`aggressive factors that act to injure the gastroduodenal
`mucosa and defensive factors that normally protect
`against corrosive agents is also important. When this
`delicate balance is disrupted for any reason, mucosal
`injury may ensue.1 These defensive properties appear to
`be mediated to a large extent by endogenous prostaglan-
`dins, nitric oxide, and trefoil proteins, and when the
`
`acid plays an etiologic role in producing symptoms and
`causing mucosal injury, such as gastroesophageal reflux
`disease (GERD). Although patients with gastric ulcers
`(GUs) tend to have normal or reduced levels of acid
`secretion,1 the average DU patient is an acid hypersecre-
`tor. When compared with age-matched controls, DU
`patients secrete ,70% more acid during the day (meal-
`stimulated) and about 150% more acid at night (basal
`secretion) (Figure 2).11 Postprandial gastric acid secretion
`is regulated primarily by increases in gastrin expression,
`which is controlled by a negative feedback loop. Individu-
`als infected with Helicobacter pylori have been shown to
`have a diminished number of somatostatin-secreting D
`
`Figure 2. Gastric acid secretion during the day and night in patients
`with DU (n 5 8) and in age-matched controls (n 5 7). Acid secretion is
`expressed as the mean 6 SE in millimoles per 12 hours. *P, 0.05.
`Data from Feldman et al.11
`
`DRL EXHIBIT 1014 PAGE 2
`
`

`

`February Supplement 2000
`
`ACID SUPPRESSION THERAPY S11
`
`synthesis of any or all are diminished, the ability of
`the gastroduodenal mucosa to resist injury is decreased.
`Thus, even normal rates of acid secretion may be
`sufficient to injure the mucosa and produce gastroduode-
`nal ulcers. Nevertheless, even in DU patients who are
`normal secretors of acid, a reduction in the rate of acid
`secretion is the most efficient means of healing ulcers.1
`Although a large number of gastroduodenal ulcers are
`associated with H. pylori
`infection, at least 60% of
`individuals with complicated ulcers (e.g., hemorrhage or
`perforation) report the use of nonsteroidal anti-inflamma-
`tory drugs (NSAIDs), including aspirin.16 Mucosal injury
`associated with NSAID use is initiated topically by the
`acidic nature of NSAIDs.17 Topical mucosal injury may
`also occur as a result of indirect mechanisms, mediated
`through the biliary excretion and subsequent duodenogas-
`tric reflux of active NSAID metabolites.18,19 Topical
`injury caused by NSAIDs certainly contributes signifi-
`cantly to the development of gastroduodenal mucosal
`injury, but the systemic effects of these agents appear to
`play the predominant role,17,20,21 largely through the
`decreased synthesis of mucosal prostaglandins.22 Avoid-
`ance of topical mucosal injury by enteric-coated aspirin
`preparations22 or by the parenteral23 or rectal24 adminis-
`tration of NSAIDs does not prevent the development of
`ulcer complications. Moreover, doses of aspirin as low as
`10 mg are sufficient to significantly suppress gastric
`mucosal prostaglandin synthesis.25 A decrease in the
`above protective mechanisms normally stimulated by
`prostaglandins enables endogenous gastric acid to incite
`mucosal injury.
`In recent years, it has become evident that the actual
`percentage of ulcers associated with H. pylori may not be
`90%–95% as often reported, but may be as low as 32% in
`non–referral-based populations.26 Furthermore, despite
`the inclination to ascribe the etiology of ulcers in such
`individuals to NSAIDs, the use of these agents clearly
`does not account for the balance of the cases. Finally, the
`vast majority of remaining individuals do not have the
`Zollinger–Ellison syndrome (ZES) or another unusual
`cause of gastroduodenal ulcer. Thus, while peptic ulcer-
`ation involves the participation of several factors, as first
`stated by Karl Schwarz27 in 1910: ‘‘Ohne sauren Magensaft,
`kein peptisches Geschwu¨r,’’ i.e., ‘‘No acid, no ulcer.’’ The
`erosive properties of acid continue to play a central role in
`the pathogenesis of gastroduodenal mucosal ulceration,
`and conversely, acid suppression therapy remains the
`cornerstone of therapy.
`
`GERD
`Although the principal aggressive factor involved
`in causing heartburn and the other clinical manifestations
`
`of GERD is the presence of acid in the esophagus, the
`disorder does not usually result from the hypersecretion
`of gastric acid.28 Rather, GERD occurs as a result of
`several abnormalities in motor function of the lower
`esophagus and the lower esophageal sphincter (LES).
`Despite the etiologic role played by these important
`motor abnormalities, the severity of symptoms, most
`notably heartburn, and esophageal mucosal injury can be
`correlated with the total time that the esophageal mucosa
`is exposed to acid. Gastric acid thus also constitutes a
`critical element in the pathogenesis of GERD, and acid
`suppression comprises
`the principal mechanism for
`therapy. However, the optimal timing and degree of acid
`suppression differ significantly in GERD patients com-
`pared with the treatment of gastroduodenal ulcer (see
`below).
`
`Stress-Related Erosive Syndrome
`Many terms have been used to describe this entity,
`including stress ulcer syndrome, stress gastritis, stress-
`related mucosal disease, and stress-related erosive syn-
`drome (SRES).29,30 The principal feature of SRES is its
`relationship to serious systemic disease, such as sepsis,
`massive burn injury, head injury associated with in-
`creased intracranial pressure, severe trauma, and multiple-
`system organ failure. A meta-analysis of 2252 patients by
`Cook et al.31 identified mechanical ventilation and
`coagulopathy as the 2 singlemost important risk factors.
`Although the pathophysiology is multifactorial and
`definitely includes a component of
`ischemia, which
`compromises gastric mucosal
`integrity,
`luminal acid
`plays a dominant role in producing the multiple erosive
`lesions characteristic of the entity. Fiddian-Green et al.32
`emphasized the importance of H1 ion back-diffusion by
`demonstrating a high correlation between the degree of
`intramural pH and the development of SRES. Further-
`more, most, but not all, methods for preventing massive
`hemorrhage-associated SRES include the alkalinization of
`gastric contents.33
`
`Pharmacology of Parietal Cell
`Receptors
`The parietal cell possesses a unique morphology
`that differs markedly between the resting and stimulated
`states.1 Mitochondria occupy 34% of its cell volume,
`indicative of the importance of adenosine triphosphate
`(ATP) synthesis as an energy source required for the active
`transport of H1 ions out of the cell against a 3,000,000:1
`ionic gradient. A large percentage of resting cell volume
`is also occupied by tubulovesicles, which are elongated
`tubes with smooth surface membranes, and by the
`
`DRL EXHIBIT 1014 PAGE 3
`
`

`

`S12 WOLFE AND SACHS
`
`GASTROENTEROLOGY Vol. 118, No. 2
`
`Table 1. Comparison of the Histamine H2-Receptor
`Antagonists
`Cimetidine Ranitidine Famotidine Nizatidine
`80
`50
`40
`70
`1
`5–10
`32
`5–10
`1.5–2.3
`1.6–2.4
`2.5–4
`1.1–1.6
`6
`8
`12
`8
`
`Bioavailability (%)
`Relative potency
`Circulator y t1⁄2 (h)
`a (h)
`Biological t1⁄2
`Relative effect on
`cytochrome P450
`metabolism
`
`1
`
`0.1
`
`0
`
`0
`
`t1⁄2, half-life.
`aApproximate values.
`Adapted and reprinted with permission. 37
`
`safety record, and in 1995 became available for use in the
`United States without prescription.
`
`Muscarinic Receptor and Its Antagonists
`The central nervous system, particularly via the
`vagus nerve, plays a dominant role in regulating basal
`acid secretion, as well as the cephalic phase of meal-
`stimulated acid secretion. Extracts of belladonna were
`used to treat dyspepsia since the time of the Roman
`empire, and in the recent past, nonspecific antimuscarinic
`agents such as atropine and propantheline bromide were
`used as inhibitors of gastric acid secretion. These drugs
`were associated with many adverse effects, including
`drowsiness, dry mouth, blurry vision, and urinary reten-
`tion, and as a result are rarely used today. To date,
`however, 5 muscarinic receptors have been subtyped and
`cloned, and although all are G protein coupled, they
`signal different intracellular pathways. In vitro character-
`ization of gastric acid secretion has indicated that the
`parietal cell normally expresses the M3 subtype.38 Clini-
`cally, the M1 antagonists, pirenzepine and telenzepine,
`are effective inhibitors of acid secretion and probably
`
`secretory canaliculus, a small invaginated area of the
`apical membrane. Upon stimulation, which is generally
`accomplished by eating a meal, the tubulovesicles de-
`crease in number and become transformed into microvilli
`around the secretory canaliculus, which serves to greatly
`expand the surface area of the parietal cell in preparation
`for the secretion of large quantities of HCl. The parietal
`cell also possesses several different receptors for stimula-
`tory and inhibitory ligands on its basolateral membrane
`(Figure 1).
`Histamine H2 Receptor and Its Antagonists
`The histamine receptor belongs to a large family
`of G protein–linked receptors possessing 7 transmem-
`brane domains.34 Despite the recognition that histamine
`stimulates gastric acid secretion, it was not until 1966,
`when Ash and Schield35 described H1 and H2 receptors
`for histamine, that the possibility of inhibiting acid
`secretion with histamine antagonists was proposed. In
`1970, Black et al.36 described selective histamine H2-
`receptor inhibition and initiated the search for pharmaco-
`logical agents that could effectively suppress the secretion
`of acid. Within 10 years of the release of cimetidine in the
`United States in 1977, 3 additional H2-receptor antago-
`nists—ranitidine, famotidine, and nizatidine—became
`available for use throughout the world. All 4 drugs
`(Figure 3) suppress basal and meal-stimulated acid
`secretion, albeit to a lesser degree than proton pump
`inhibitors (PPIs) discussed below. Despite similar thera-
`peutic profiles, some differences do exist with regard to
`the agents’ pharmacokinetic properties (Table 1), most of
`which are clinically insignificant.37 The elimination of
`these drugs occurs by a combination of hepatic metabo-
`lism and urinary excretion, and although hepatic dysfunc-
`tion does not alter their pharmacokinetic properties, dose
`reductions are recommended for all individuals with
`varying degrees of renal impairment (Table 2).37 H2-
`receptor antagonists as a class possess an unsurpassed
`
`Drug
`Cimetidine
`
`Ranitidine
`
`Famotidine
`
`Nizatidine
`
`Table 2. Histamine H2-Receptor Antagonist Dosing
`Adjustments With Renal Insuf ficiency
`Creatinine clearance (mL/min)
`.30
`15–30
`,15
`.75
`30–75
`15–30
`,15
`.75
`30–75
`15–30
`,15
`.75
`30–75
`15–30
`,15
`
`Dose (mg/day)a
`800
`600
`400
`300
`225
`150
`75
`40
`30
`20
`10
`300
`225
`150
`75
`
`Figure 3. Structure of the 4 H2-receptor antagonists in use in the
`United States.
`
`aDosing for gastroduodenal ulcer.
`Adapted and reprinted with permission. 37
`
`DRL EXHIBIT 1014 PAGE 4
`
`

`

`February Supplement 2000
`
`ACID SUPPRESSION THERAPY S13
`
`exert their effect by interaction with a postsynaptic
`neuronal M1 receptor.1 These 2 agents are available in
`other countries for the treatment of duodenal ulcer, but
`they have not been approved for use in the United States.
`
`Gastrin Receptor and Its Antagonists
`As stated above, gastrin stimulates acid secretion
`via an endocrine pathway and induces H1 ion generation
`both directly and indirectly. The precise location and
`density of the gastrin receptor have been subjects of
`debate and may be somewhat species dependent. Several
`studies in rats and other rodents have suggested that
`gastrin stimulates acid secretion by enhancing the release
`of histamine from ECL cells.39,40 Conversely, receptors for
`gastrin receptors on canine parietal cells have been
`demonstrated by Soll et al.41 by employing the analogue
`125I-[Leu15]gastrin, and by Kopin et al.,42 who cloned and
`expressed the gastrin receptor from a purified canine
`parietal cell complementary DNA library. Moreover,
`single-cell video imaging has provided direct evidence for
`a functional gastrin receptor on the parietal cells of rats
`and rabbits.43
`The gastrin receptor belongs to the above-mentioned
`family of G protein–linked receptors possessing 7 trans-
`membrane domains.42 It is closely related to the receptor
`for cholecystokinin (‘‘CCK-A’’ or ‘‘CCK-1’’) and is thus
`often referred to as the ‘‘CCK-B’’ or ‘‘CCK-2’’ receptor.
`Gastrin-specific receptor antagonists have been devel-
`oped, which include L365,260 and YM022.44 The former
`is a benzodiazepine derivative that was derived from the
`fungus Aspergillus alliaceus and has been demonstrated to
`effectively antagonize gastrin-stimulated gastric acid
`secretion.44 Despite this effect, these antagonists have not
`been used clinically as inhibitors of acid secretion.
`However, they may ultimately prove useful
`in the
`treatment of panic and anxiety disorders by virtue of
`binding to gastrin receptors in the brain.45
`
`Miscellaneous Receptors on the
`Parietal Cell
`Other receptors on the parietal cell basolateral
`membrane have been suggested by the ability of various
`agents to inhibit gastric acid secretion. For example,
`prostaglandins inhibit H1 ion generation by binding to
`their EP3 G protein–linked receptor on the parietal cell,
`which appears to inhibit adenylate cyclase, and thereby
`decrease intracellular cAMP generation when activated.46
`As discussed below, the acid-inhibitory properties of
`prostaglandin analogues such as misoprostol, while not
`potent, are nevertheless critical for exerting any beneficial
`clinical effects.16
`
`Parietal Cell H11,K11-Adenosine
`Triphosphatase
`The gastric enzyme H1,K1-adenosine triphospha-
`tase (ATPase) is a member of the family of ion-motive
`ATPases that includes F1, V, and P ATPases. The latter is
`divided conveniently into P1 or P2 types in 1 of 2 ways,
`either based on the number of transmembrane segments
`(8 in the case of the P1 and 10 in the case of the P2
`catalytic subunits) or based on transport of transition
`metals (P1) or small cations (P2).47 Within the P2 family,
`the gastric H1,K1- and Na1,K1-ATPase isoforms are
`coexpressed tightly bound to a b-subunit that is smaller
`than the catalytic or a-subunit, has most of its sequence
`presented outside, and is glycosylated to different extents
`depending on the isoform.48,49 These latter 2 enzymes are
`also unique in that they both serve as drug targets,
`digoxin for congestive heart failure in the case of the
`Na1,K1-ATPase and substituted pyridyl methylsulfinyl
`benzimidazoles for acid-related disorders in the case of
`the H1,K1-ATPase.50 H1,K1-ATPase consists of 8 trans-
`membrane segments for the catalytic a-subunit and 1
`transmembrane segment for the b-subunit (Figure 4).51,52
`
`Inhibition of Acid Secretion
`The recognition that the ATPase was the final step
`of acid secretion has resulted in the development of a class
`of drugs, the PPIs, that are targeted toward this enzyme.
`They all share a common structural motif (Figure 5), a
`substituted pyridylmethylsulfinyl benzimidazole, but vary
`in terms of their substitutions. They also share common
`inhibitory mechanisms and are all weak protonatable
`pyridines, with a pKa of ,4.0 for omeprazole, lansopra-
`zole, and pantoprazole, and ,5.0 for rabeprazole. They
`thus accumulate selectively in the acid space of the
`secreting parietal cell. Within that space or on the surface
`
`Figure 4. A model of the 2-dimensional arrangement of the gastric
`H1,K1-ATPase subunits. The core structure of this P2-type ATPase
`consists of the first 6 membrane segments, arranged as helices with
`the possible exception of the cytoplasmic end of M6. Cross-linking and
`column chromatography of tryptic fragments has shown an associa-
`tion between M5 and M7, and M6 and M9 (curvedarrows). The major
`high-affinity interaction with the b-subunit is with the beginning of M8.
`
`DRL EXHIBIT 1014 PAGE 5
`
`

`

`S14 WOLFE AND SACHS
`
`GASTROENTEROLOGY Vol. 118, No. 2
`
`Figure 5. Structure and reaction pathways of the 4 PPIs in use in the United States. All are ampholytic weak bases and accumulate as prodrugs in
`the secretor y canaliculus of the actively secreting parietal cell. They are all acid-catalyzed to a reactive thiophilic species whereby the pyridine N
`reacts with the 2C of the benzimidazole to form a sulfenic acid (below), which, while in solution, reacts further to form a sulfenamide (above). The
`respective sulfenamides then react with available cysteines on the luminal sur face of the enzyme; cys 813 is common to all the PPIs and crucial
`for enzyme inhibition. Cys 892 reacts with omeprazole,
`lansoprazole, and rabeprazole; cys 321 with lansoprazole; and cys 822 with
`pantoprazole. 54
`
`of the enzyme, they undergo an acid-catalyzed conversion
`to a reactive species, the thiophilic sulfenamide or
`sulfenic acid, which are permanent cations. The rate of
`conversion varies among the compounds and is a function
`of their pKa and other structural features: rabeprazole .
`omeprazole 5 lansoprazole . pantoprazole.53,54 The
`reactive species reacts with cysteine residues available
`from the external surface of the enzyme that faces the
`lumen of the secretory space of the parietal cell, resulting
`in covalent inhibition of the enzyme by disulfide bond
`formation. The cysteine residue that is critical
`for
`inhibition is cys 813 in the catalytic subunit of the
`H1,K1-ATPase, strategically located in the M5/M6
`sector of the enzyme, which is intimately involved in the
`transport process.
`Because all the PPIs require accumulation and acid
`activation, their onset of
`inhibition is delayed, and
`because they also form a covalent derivative, the restora-
`tion of acid secretion is likewise delayed, depending on
`the turnover of the pump protein and the biological
`reversibility of the disulfide bond. Therefore, 24–48
`hours are necessary for maximal acid secretory capacity to
`be restored. Different doses of these drugs are recom-
`mended, but at equivalent doses, these agents are remark-
`ably similar when used in the treatment of acid-related
`disorders (Table 3).55
`The PPIs are without question the most potent
`inhibitors of gastric acid secretion available. However,
`because they are most effective when the parietal cell is
`stimulated to secrete acid in response to a meal, these
`
`drugs should only be taken before or with a meal and
`should not be used in conjunction with H2-receptor
`antagonists, prostaglandins, or other antisecretory agents
`(Table 3). Animal studies have demonstrated that the
`concomitant administration of PPIs and other antisecre-
`tory agents will markedly reduce the acid-inhibitory
`effects of the PPI.56 Because acid secretion must be
`stimulated for maximum efficacy, PPIs are administered
`before the first meal of the day (Table 3). Moreover, in
`
`Table 3. Helpful Facts on the Use of PPIs
`PPIs are prodrugs that require activation to their active moiety (thio-
`philic sulfenamide)
`pKa of PPIs: ,4 for omeprazole, lansoprazole, and pantoprazole; ,5
`for rabeprazole
`All PPIs are activated when regional pH decreases below their respec-
`tive pKa
`Achieved almost exclusively in parietal cell secretor y canaliculus
`Achieved optimally when parietal cell is activated, i.e., after meals
`Most effective after a prolonged fast, when a large amount of inac-
`tive H1,K1-ATPase present is in secretor y canaliculus, i.e.,
`after breakfast
`Clinical use
`Steady state not achieved for several days
`Thus often helpful to administer twice daily for the first 2–3 days
`of therapy
`Also likely not to be consistently clinically effective when taken
`sporadically
`First dose should be taken before breakfast
`Second dose, if used, should be taken before evening meal
`Donotadminister concomitantly with H2-receptor antagonists or
`prostaglandins
`Safety: toxicity seems to be gastrin-related and is probably species
`specific
`
`DRL EXHIBIT 1014 PAGE 6
`
`

`

`February Supplement 2000
`
`ACID SUPPRESSION THERAPY S15
`
`most individuals, once-daily dosing is sufficient to
`produce the desired level of acid inhibition. A second
`dose, if required, should be administered before the
`evening meal.
`During meals, however, neither all parietal cells nor all
`proton pumps are active. The initial dose of the PPI will
`thus inhibit only activated H1,K1-ATPase present in the
`canalicular membrane.56 As inactive enzyme is recruited
`into the secretory canaliculus, acid secretion will resume,
`albeit at a reduced level. After a second dose, more
`enzyme will have been recruited and subsequently inhib-
`ited, and after a third dose, additional recruitment and
`further acid inhibition may be expected. Once-daily PPI
`dosing results in 66% steady-state inhibition of maximal
`acid output after 5 days, and the initial use of twice-daily
`dosing (for the first 2–3 days) may thus be helpful in
`achieving more rapid inhibition of gastric acid secre-
`tion.55 Based on these pharmacokinetic properties, the
`occasional use of a PPI taken on an ‘‘as-needed’’ basis
`would not be expected to provide adequate acid inhibi-
`tion and would be unlikely to produce a consistent or
`satisfactory clinical response.
`
`Safety Issues
`The safety profile of the PPIs is similar to that of
`H2-receptor antagonists. Although headache and diarrhea
`are occasionally reported, serious adverse reactions to
`these agents are rare and generally not well documented.
`Initial concern was expressed when PPIs were introduced
`as a result of their ability to precipitate ECL cell
`hyperplasia and subsequent gastric carcinoid tumors in
`rats when given at high doses. This trophic effect occurs
`as a result of
`interruption of the negative feedback
`mechanism for acid secretion, which induces an increase
`in antral gastrin gene expression.1,4–6 However, these
`drugs have been used for nearly 15 years in Europe and for
`10 years in the United States with no discernible increase
`in the incidence of carcinoid tumors. The reasons for such
`interspecies differences are not entirely evident, but are
`probably related in part to a lower mucosal ECL cell
`density and a considerably less pronounced increase in
`circulating gastrin in humans compared with rodents in
`response to antisecretory medications. One area of theo-
`retical concern that has not been completely resolved is
`the possible effect of gastrin and its precursor prohor-
`mone on promoting the growth of colonic neoplasms.
`An area of controversy has been the need to monitor
`serum gastrin concentrations in persons on long-term
`PPI therapy. In general, laboratory and other testing
`should be performed only if the treatment plan will be
`dependent on their results. Long-term PPI treatment is
`used most commonly in patients with GERD, many of
`
`whom have significant symptoms and mucosal injury.
`The sequelae of GERD are serious and real, whereas the
`effects of hypergastrinemia, which occurs in only ,5% of
`individuals receiving long-term PPI therapy, are theoreti-
`cal. Therefore, in situations in which the hypothetical
`risk of the PPI is clearly outweighed by the benefits
`offered by potent acid suppression, serum gastrin measure-
`ments should not be performed.57
`
`Treatment of Acid-Related Disorders
`Acute Gastroduodenal Ulcer
`The treatment of ulcer disease has changed dra-
`matically since the discovery that the probability of ulcer
`recurrence decreases significantly after successful eradica-
`tion of H. pylori infection, compared with annual recur-
`rence rates of 50%–80% when antisecretory therapy
`alone is used.58,59 Thus, a determination of H. pylori
`infection in a patient with gastroduodenal ulcer is critical
`for the appropriate management of this disease. If a
`patient is not infected with H. pylori, an alternative
`etiology must be sought, such as NSAID use, hypersecre-
`tory states, or one of the other less common causes of ulcer
`disease, such as Crohn’s disease, vascular insufficiency,
`viral infection, radiation therapy, and cancer chemother-
`apy. Regardless of the etiology, however, the inhibition of
`gastric acid secretion continues to play a prominent role
`in the management of acute gastroduodenal ulcer.
`Some differences do exist between DUs and GUs
`proximal to the prepyloric region of the stomach. Most
`previous studies have assessed the effects of acid suppres-
`sion on duodenal and pyloric channel ulcers and antral
`ulcers within 2–3 cm of the pylorus, but few have
`actually evaluated the healing of more proximal GUs.
`Although gastric acid secretion is generally lower than
`normal in patients with more proximal disease,1 these
`latter ulcers usually do heal in response to acid suppres-
`sion, although the total duration of therapy is often
`prolonged.
`In general, a dynamic relationship exists between the
`healing of DU and the inhibition of intragastric acidity.
`Important parameters that determine the effect of antise-
`cretory therapy include the degree of suppression of
`intragastric acidity, the length of acid inhibition over a
`24-hour period, and the duration of antisecretory treat-
`ment. For example, Burget et al.60 showed that if
`intragastric pH is maintained above 3.0 for a period of
`18–20 hours per day, DU healing approximates 100% at
`4 weeks. As discussed below, the DU healing rate at 4
`weeks is also directly proportional to the degree of the
`reduction of nocturnal acidity.61
`
`DRL EXHIBIT 1014 PAGE 7
`
`

`

`S16 WOLFE AND SACHS
`
`GASTROENTEROLOGY Vol. 118, No. 2
`
`Table 4. Current Recommendations for Treatment of
`Gastroduodenal Ulcers
`Active ulcer
`H2-receptor antagonists
`Cimetidine 800 mg
`Ranitidine/nizatidine 300 mg
`Famotidine 40 mg
`All administer ed between the evening meal and bedtime
`PPIs
`Omeprazole 20 mg
`Lansoprazole 30 mg
`Rabeprazole 20 mg
`Pantoprazole 40 mg
`All administer ed daily before breakfast
`Maintenance therapy
`H2-receptor antagonists
`Cimetidine 400 mg
`Ranitidine/nizatidine 150 mg
`Famotidine 20 mg
`All administer ed between the evening meal and bedtime
`PPIs
`As above
`Prevention of NSAID-induced ulcers
`Misoprostol
`At least 200 µg 3 times/day
`PPIs
`As above
`
`NOTE. In general, duodenal ulcers should be treated for 4 weeks and
`gastric ulcers for 8 weeks.
`
`Antacids. In a landmark controlled, double-blind
`study in 1977, Peterson et al.62 showed that antacids were
`superior to placebo in healing DU, although (contrary to
`prevailing opinions) no difference in symptom relief was
`detected. However, because of the need to take these
`d

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket