throbber
144
`
`WALLACE ET AL.
`DRUG DEVELOPMENT RESEARCH 42:144–149 (1997)
`
`Research Overview
`Gastrointestinal-Sparing Anti-Inflammatory Drugs:
`The Development of Nitric Oxide-Releasing NSAIDs
`John L. Wallace,1* Susan N. Elliott,1 Piero Del Soldato,2 Webb McKnight,1
`Franco Sannicolo,3 and Giuseppe Cirino4
`1Department of Pharmacology, The University of Calgary, Calgary, Alberta, Canada
`2NicOx Ltd., Paris, France
`3Department of Organic Chemistry, University of Milan, Milan, Italy
`4Department of Experimental Pharmacology, University of Naples, Naples, Italy
`
`Strategy, Management and Health Policy
`
`Venture Capital
`Enabling
`Technology
`
`Preclinical
`Research
`
`Preclinical Development
`Toxicology, Formulation
`Drug Delivery,
`Pharmacokinetics
`
`Clinical Development
`Phases I-III
`Regulatory, Quality,
`Manufacturing
`
`Postmarketing
`Phase IV
`
`Nonsteroidal anti-inflammatory drugs (NSAIDs) are among the most widely prescribed medi-
`ABSTRACT
`cations, but their use continues to be limited by significant toxicity, particularly in the gastrointestinal tract
`and kidney. Better understanding of the pathogenesis of these adverse effects has led to the development of
`a series of derivatives of standard NSAIDs that are not only less toxic but more efficacious. The coupling of
`a nitric oxide-releasing moiety to a range of NSAIDs greatly reduces their ability to induce gastrointestinal
`damage, and greatly increases their tolerability in situations in which there is preexisting gastrointestinal
`inflammation. There is also evidence that these compounds are much better tolerated by the kidney. On the
`other hand, the analgesic and anti-thrombotic properties of NO-releasing NSAIDs significantly exceed those
`of the parent drugs. These compounds appear to represent a significant advance in the treatment of
`inflammation and pain and for prophylaxis of thrombotic conditions. Drug Dev. Res. 42:144–149, 1997.
`© 1997 Wiley-Liss, Inc.
`
`Key words:
`
`anti-inflammatory; ulcer; analgesic; anti-thrombotic
`
`INTRODUCTION
`The class of drugs known as ‘‘NSAIDs’’ (nonsteroi-
`dal anti-inflammatory drugs) are among the most com-
`monly used medications [Garner, 1992]. They are
`prescribed largely for their anti-inflammatory, antipyretic,
`and analgesic properties but are also very widely used in
`over-the-counter preparations for the same indications.
`Moreover, aspirin is increasingly used on a long-term
`basis for its well-documented anti-thrombotic effects. The
`major limitation to the use of NSAIDs is their ability to
`cause ulceration and bleeding in the gastrointestinal
`tract [Soll et al., 1991]. This effect of NSAIDs has been
`recognized for decades [Douthwaite and Lintott,
`1938]. More recently, it has become apparent that
`NSAIDs can also exert significant toxicity in the kid-
`ney [Segasothy et al., 1994].
`Over the past 40 years, numerous new NSAIDs
`
`© 1997 Wiley-Liss, Inc.
`
`have been marketed with the claim that they reduce gas-
`trointestinal toxicity. These approaches have included
`enteric coating of the drug to prevent absorption in the
`stomach, formulation as a pro-drug, to prevent contact
`between the active drug and the gastric mucosa, and de-
`livery by non-oral routes. The basis for these approaches
`was the observation that some NSAIDs, particularly those
`which are acidic, can directly damage gastric epithelial
`cells. Reducing the topical irritancy should therefore pre-
`vent the epithelial damage and, in turn, prevent ulcer-
`ation and bleeding. However, each of these approaches
`has failed in terms of reducing the truly significant ad-
`
`*Correspondence to: Dr. John L. Wallace, Department of Phar-
`macology and Therapeutics, The University of Calgary, 3330 Hospi-
`tal Drive, NW, Calgary, AB T2N 4N1, Canada. E-mail:wallacej@
`acs.ucalgary.ca
`
`Page 1 of 6
`
`Patent Owner Ex. 2063
`Mylan v. Pozen
`IPR2017-01995
`
`

`

`NO-RELEASING NSAIDS
`
`145
`
`verse effects of NSAIDs in the gastrointestinal tract [Gra-
`ham, 1990]. This is now recognized as being attributable
`to the fact that a central component of the mechanism
`underlying NSAID-induced ulceration and bleeding is
`their ability to inhibit prostaglandin synthesis in the gas-
`trointestinal mucosa. Since the desired effects of NSAIDs
`are also dependent upon suppression of prostaglandin
`synthesis [Vane, 1971], delivery of the drug to the sys-
`temic circulation at a dose that is effective invariably re-
`sulted, in some patients, in development of ulcers.
`
`WHY NITRIC OXIDE?
`NSAIDs inhibit prostaglandin synthesis by inhib-
`iting the activity of the enzyme cyclo-oxygenase. Pros-
`taglandins play an important role in the gastrointestinal
`tract in that they mediate several components of mucosal
`defense (blood flow, mucus and bicarbonate secretion,
`mucosal immunocyte function) [Wallace and Tigley, 1995].
`Inhibition of gastrointestinal prostaglandin synthesis
`leads to decreased mucus and bicarbonate secretion,
`decreased blood flow, impaired repair of epithelial in-
`jury, and an increase in the number of leukocytes adher-
`ing to the vascular endothelium in the gastrointestinal
`microcirculation. It is this latter observation [Wallace,
`1993] that first suggested that nitric oxide (NO) might be
`capable of preventing NSAID-induced gastric injury. NO
`can inhibit leukocyte adherence to the vascular endot-
`helium [Kubes et al., 1991]. We had previously observed
`that prevention of NSAID-induced leukocyte adherence
`resulted in near-complete protection against gastric in-
`jury associated with these drugs in animals [Wallace et
`al., 1990, 1991, 1993]. Moreover, NO also exhibits many
`of the same actions in the stomach as prostaglandins;
`namely, stimulation of mucus secretion [Brown et al.,
`1992] and maintenance of mucosal blood flow [Whittle,
`1993]. Thus, it seemed logical that if NO could be deliv-
`ered in a controlled manner (i.e., slowly, so that systemic
`arterial blood pressure would not be affected), the detri-
`mental effects of NSAIDs in the gastrointestinal tract
`might be prevented (Fig. 1). The rate of delivery of NO,
`in terms of effectiveness in preventing NSAID-induced
`gastric damage, was underscored by our observations that
`some doses of standard NO donors (glyceryl trinitrate,
`sodium nitroprusside) were capable of reducing the ex-
`tent of gastric damage induced by an NSAID [Wallace et
`al., 1994c], but small increases in the dose led to an exac-
`erbation of injury (unpublished observations). Moreover,
`it was difficult to identify doses of these drugs which were
`effective in preventing gastric injury without altering
`systemic blood pressure. Nevertheless, the concept that
`NO was capable of reducing the severity of NSAID-in-
`duced gastric injury was proven [Wallace et al., 1994c].
`Protective effects of NO in other models of gastric injury
`had previously been demonstrated [MacNaughton et al.,
`
`Schematic diagram illustrating the rationale behind the develop-
`Fig. 1.
`ment of NO-releasing NSAIDs. As the compounds retain the ability to
`inhibit cyclo-oxygenase, they inhibit prostaglandin synthesis as effectively
`as the parent drugs. However, the release of NO counteracts the detri-
`mental effects of inhibition of prostaglandin synthesis in the gastrointesti-
`nal tract. The release of NO also appears to enhance the analgesic and
`anti-thrombotic efficacy of the drugs over that of the parent drugs.
`
`1989], but again, increasing the dose of the NO donor
`led to an exacerbation of injury and profound effects on
`systemic blood pressure [Lopez-Belmonte et al., 1993].
`
`NO-NSAIDS: REDUCED TOXICITY
`The hypothesis that NO-releasing derivatives of
`standard NSAIDs would have reduced toxicity has now
`been tested using derivatives of flurbiprofen, ketoprofen,
`naproxen, aspirin, and diclofenac [Wallace et al., 1994a,b,
`1995a, 1997; Davies et al., 1996]. When given as a single
`dose, all of the NO-NSAIDs have been shown to pro-
`duce significantly less gastric damage than standard
`NSAIDs [Wallace et al., 1994a,b, 1995a; Davies et al.,
`1996]. Figure 2 shows the effects of an NO-aspirin de-
`rivative (NCX-4016) in comparison to aspirin on the rat
`stomach. A similar reduction in toxicity is apparent if the
`compounds are given systemically rather than orally, in-
`dicating that the reduced injury observed with these com-
`pounds is not solely due to reduced ”topical irritant“
`properties. With repeated administration for up to three
`weeks, these compounds were also shown to produce sig-
`nificantly less gastrointestinal injury [Wallace et al., 1994a;
`Reuter et al., 1994; Davies et al., 1996].
`A major clinical problem associated with the use of
`NSAIDs is the ability of these drugs to interfere with the
`healing of preexisting ulcers. For this reason, we assessed
`the effects of NO-NSAIDs in three models. First, we stud-
`ied the effects of an NO-NSAID in a model of colitis in
`the rat [Reuter et al., 1994]. In this model, standard
`
`Page 2 of 6
`
`Patent Owner Ex. 2063
`Mylan v. Pozen
`IPR2017-01995
`
`

`

`146
`
`WALLACE ET AL.
`
`Extent of gastric damage induced in the rat stomach 3 h after
`Fig. 2.
`oral administration of aspirin or equimolar doses of an NO-aspirin de-
`rivative (NCX-4016). Aspirin caused significant damage at doses of 30
`mg/kg or greater (u = P < 0.05, uu = P < 0.01 vs. the vehicle-treated
`group), while NCX-4016 did not cause damage at any dose tested.
`
`NSAIDs cause a marked exacerbation of colonic injury,
`leading to perforation and death. For example, in the case
`of diclofenac, treatment for one week at doses of 1, 5,
`and 10 mg/kg led to the death of 29%, 86%, and 100% of
`the rats, respectively. In contrast, treatment with equimo-
`lar doses of nitrofenac resulted in markedly less mortal-
`ity (0%, 0%, and 33%, respectively). The second model
`was one in which the effect of nitrofenac on healing of
`gastric ulcers in the rat were examined [Elliott et al.,
`1995]. While daily diclofenac administration for a week
`caused a significant reduction in body weight gain and a
`marked decrease in hematocrit, nitrofenac did not alter
`either of these parameters in comparison to the vehicle-
`treated control group. Moreover, nitrofenac actually ac-
`celerated ulcer healing. The third model we utilized was
`one in which intestinal injury is established by treatment
`with indomethacin, and the rats are then switched to a
`different NSAID, to the corresponding NO-NSAID, or
`to vehicle. This model mimics the clinical situation in
`which a patient develops gastrointestinal damage while
`taking an NSAID, and has to be switched to a second
`NSAID in the hope that it will be less injurious. The re-
`sults from one study performed with this model are shown
`in Figure 3. Intestinal injury was induced by two subcu-
`taneous injections of indomethacin (7.5 mg/kg) 24 h apart.
`The rats were left for 48 h, after which they were ran-
`domized to receive naproxen (20 mg/kg), an equimolar
`dose of NO-naproxen, or vehicle. The test drugs or ve-
`hicle were administered orally every 12 h for 4 days, af-
`ter which time the degree of intestinal ulceration was
`blindly scored. Indomethacin induced widespread intes-
`tinal ulceration. Considerable healing was observed in
`the group subsequently treated with vehicle. In contrast,
`
`Effects of naproxen and NO-naproxen on healing of intestinal
`Fig. 3.
`ulcers induced by indomethacin. Indomethacin was injected subcutane-
`ously (7.5 mg/kg) twice, separated by 24 h. 48 h after the second in-
`domethacin injection (day 3), the rats were randomized to receive
`naproxen, an equimolar dose of NO-naproxen, or vehicle. These drugs
`were given orally every 12 h for 4 days. 12 h after the final dose, the
`damage was scored. * = P < 0.05 compared to the vehicle-treated group.
`
`naproxen significantly delayed healing. On the other
`hand, NO-naproxen, which suppresses prostaglandin
`synthesis in the intestine to the same extent as naproxen,
`did not cause any retardation of ulcer healing.
`The fact that gastrointestinal toxicity was not ob-
`served following repeated administration of the NO-
`NSAIDs over periods of up to three weeks further
`supports the contention that these derivatives are not
`merely pro-drugs. Moreover, recent pharmacokinetic
`studies have demonstrated that the intact NO-NSAIDs
`can be detected in the plasma of rats for many hours af-
`ter administration [Davies et al., 1996; Reuter et al., 1997].
`Despite evidence that these agents generate NO in the
`rat in vivo, they do not significantly affect systemic blood
`pressure [Wallace et al., 1994a,b, 1995a].
`There is also evidence suggesting that NO-NSAIDs
`spare the renal system. We tested the effects of administra-
`tion of an NSAID (diclofenac) or the NO-releasing deriva-
`tive (nitrofenac) on renal blood flow in both healthy and
`cirrhotic rats. In both subgroups, diclofenac caused a sig-
`nificant reduction in renal blood flow, while nitrofenac had
`no effect. The effects of the two drugs on renal blood flow
`in healthy rats are shown in Figure 4. As the renal toxicity
`of NSAIDs is believed to be attributable to reduced blood
`flow secondary to suppression of prostaglandin synthesis
`[Segasothy et al., 1994], these results suggest that NO-
`NSAIDs may not produce the renal toxicity associated with
`the parent NSAIDs. This conclusion is supported by re-
`cent studies in which NO-NSAIDs were found to be sig-
`nificantly better tolerated in a rat model of kidney failure
`than native NSAIDs [Fujihara et al., 1995].
`
`Page 3 of 6
`
`Patent Owner Ex. 2063
`Mylan v. Pozen
`IPR2017-01995
`
`

`

`NO-RELEASING NSAIDS
`
`147
`
`generate NO when incubated with platelets, and to
`cause a significant increase in platelet cyclic GMP
`levels [Wallace et al., 1995a, 1997a]. Indeed, there is
`strong evidence that a significant component of the
`anti-platelet effects of NO-aspirin derivatives is due
`to NO generation [Wallace et al., 1995a]. These com-
`pounds also have the ability to suppress neutrophil
`adherence to the vascular endothelium [Wallace et al.,
`1997a], which might add to their anti-thrombotic ef-
`fects. This raises the possibility that an NO-releasing
`aspirin derivative may have utility in long-term use
`for the prevention of myocardial infarction and stroke.
`Aspirin is presently used for these indications [SALT
`Collaborative Group, 1991; Meade et al., 1992;
`Patrono, 1994], but despite the fact that low doses of
`this drug are used, there is still a significant increase
`in the incidence of gastrointestinal bleeding and other
`hemostatic complications [SALT Collaborative Group,
`1991; Meade et al., 1992; Cryer et al., 1995].
`
`NITRATE VS. NITRITE: A CRITICAL DIFFERENCE
`Several options existed with respect to the NO-
`releasing moiety to link to NSAIDs. For example, nitrites,
`nitrates, and S-thiol-glutathione are all well-characterized
`NO donors [Moncada et al., 1991]. The selection of a nitrate
`group was based on two key issues: stability and toxicity.
`Stability of a flurbiprofen nitrite ester and a
`flurbiprofen nitrate ester were compared using nuclear
`magnetic resonance. When dissolved in dimethylsulfox-
`ide and incubated at 20°C for 1 h, the nitrite-containing
`compound was found to undergo 25% hydrolysis, while
`no hydrolysis of the nitrate-containing compound oc-
`curred. Moreover, the native nitrate-containing com-
`pound was found to be stable at room temperature for
`more than a year.
`In terms of toxicity, the primary concern in choos-
`ing an appropriate NO-releasing group was mutagenic-
`ity. The Ames test, which measures the potential of a test
`substance to induce point mutations in Salmonella
`typhimurium, was used to compare the effects of sodium
`nitrite, sodium nitrate, S-nitroso-glutathione, and four
`nitrate-containing NO-NSAIDs (derivatives of flurbi-
`profen, aspirin, naproxen, and diclofenac). In this assay,
`mutagenic substances induce reversion of a histidine-
`deficient strain of S. typhimurium such that it is then able
`to grow and form colonies in a histidine-limited medium.
`Concentrations up to 5000 m g per plate of each drug were
`tested. As illustrated in Figure 5, sodium nitrite caused a
`significant increase in the number of revertants (over that
`seen with vehicle) at a concentration as low as 312.5 m g/
`plate, while S-nitroso-glutathione induced a positive re-
`sponse at ‡ 1250 m g/plate. On the other hand, sodium ni-
`trate and all four of the nitrate-containing NO-NSAIDs
`
`Effects of diclofenac (10 mg/kg) and an equimolar dose of its
`Fig. 4.
`NO-releasing derivative, nitrofenac, on renal blood flow in healthy rats.
`u = P < 0.05 compared to the corresponding basal period.
`
`NO-NSAIDS: EFFICACY
`Several of the NO-NSAIDs have been compared
`to the parent drugs in models of acute and chronic in-
`flammation [Wallace et al., 1994a,b; Cuzzolin et al., 1995;
`Davies et al., 1996]. In each case, these compounds were
`found to reduce inflammation at least as effectively as
`the parent NSAID. The NO-releasing diclofenac deriva-
`tive has also been shown to have virtually identical
`anti-pyretic activity to diclofenac [Wallace et al.,
`1995b]. The NO-naproxen derivative has significantly
`enhanced analgesic activity over the parent drug
`[Davies et al., 1996], while an NO-aspirin derivative
`was found to have markedly increased anti-thrombotic
`properties [Wallace et al., 1995a].
`The ability of NO-NSAIDs to suppress inflam-
`mation, pain, and fever is not surprising given the ob-
`servation that these derivatives retain the ability of
`the parent drugs to inhibit prostaglandin synthesis
`(with the exception of the NO-aspirin derivatives; see
`below) [Wallace et al., 1994a,b; Mitchell et al., 1994].
`The NO-NSAIDs themselves inhibit cyclo-oxygenase
`activity (types 1 and 2) without being metabolized,
`indicating that these derivatives are not just pro-drugs
`[Mitchell et al., 1994]. In the case of the NO-aspirin
`derivatives, however, the activity on cyclo-oxygenase
`activity is greatly reduced, perhaps contributing to the
`profoundly reduced gastric toxicity of these drugs
`[Wallace et al., 1995a]. Both NCX-4215 and NCX-4016
`have been shown to have weak inhibitory activity on
`platelet thromboxane synthesis [Wallace et al., 1995a,
`1997b]. With repeated dosing over several days, how-
`ever, the level of inhibition of thromboxane synthesis
`increases to levels comparable to equimolar doses of
`aspirin [Wallace et al., 1997b]. However, this weak
`effect on thromboxane synthesis does not account for
`the anti-thrombotic properties of the two NO-aspirin
`derivatives. These compounds have been shown to
`
`Page 4 of 6
`
`Patent Owner Ex. 2063
`Mylan v. Pozen
`IPR2017-01995
`
`

`

`148
`
`WALLACE ET AL.
`
`Effects of sodium nitrate, sodium nitrite, and S-nitroso-glutathione
`Fig. 5.
`on mutations of S. typhimurium. Results are shown as the number of
`
`revertants per plate following exposure to the test substances at a range
`of concentrations. * = P < 0.05 compared to the vehicle-treated group.
`
`failed to induce a positive response at any of the concen-
`trations tested.
`The differences in mutagenicity of nitrate- vs. ni-
`trite-containing compounds can also be considered in the
`context of the amounts of these substances likely to be
`ingested in comparison to World Health Organization
`recommendations [JECFA, 1996]. We calculated the
`amount of nitrate and nitrite that would be ingested per
`day based on maximum daily doses of the parent NSAID.
`These calculations were performed for nitrite- and ni-
`trate-containing NO-releasing derivatives of diclofenac,
`aspirin, flurbiprofen, naproxen, and ketoprofen. In the
`case of nitrite-containing NO-NSAIDs, ingestion of any
`of the compounds would result in nitrite ingestion far in
`excess of WHO recommendations. For example, with
`NO-diclofenac, NO-aspirin, and NO-naproxen, the ni-
`trite consumption would exceed WHO recommended
`maximal levels by threefold, tenfold, and 40-fold, respec-
`tively. On the other hand, in none of the five cases exam-
`ined would the amount of nitrate ingested with
`nitrate-containing NO-NSAIDs exceed WHO recom-
`mendations. For example, the amount of nitrate ingested
`with maximal doses of NO-diclofenac and NO-flurbi-
`profen would be 1/25th and 1/7th of the WHO recom-
`mended maximal levels, respectively.
`
`SUMMARY
`NO-releasing NSAIDs were developed on the ba-
`sis of observations that suggested that NO had the ca-
`pacity to interfere with several key steps in the
`pathogenesis of NSAID-induced gastrointestinal ulcer-
`ation. This novel class of compounds exhibits greatly re-
`duced gastrointestinal and renal toxicity. NO-NSAIDs
`exhibit reduced toxicity not only in the normal gas-
`trointestinal tract, but also in situations in which there
`
`was preexisting ulceration and inflammation. Moreover,
`these compounds have comparable anti-inflammatory and
`anti-pyretic activity to the parent drugs, but have en-
`hanced analgesic and anti-thrombotic properties. Nitrate-
`containing NO-NSAIDs have superior stability to
`nitrite-containing NO-NSAIDs. Moreover, the nitrate-
`containing NO-NSAIDs do not exhibit the mutagenicity
`seen with nitrite-containing NO-NSAIDs. Clinical trials
`are presently under way to examine the safety and effi-
`cacy of several NO-NSAIDs in humans.
`
`REFERENCES
`Brown JF, Hanson PJ, Whittle BJR (1992): Nitric oxide donors increase
`mucus gel thickness in rat stomach. Eur J Pharmacol 223:101–104.
`Cryer B, Luk G, Feldman M (1995): Effects of very low doses of aspi-
`rin (ASA) on gastric, duodenal and rectal prostaglandins (PGs) and
`mucosal injury (Abstract). Gastroenterology 108:A77.
`Cuzzolin L, Conforti A, Adami A, Lussignoli S, Memestrina F, Del
`Soldato P, Benoni G (1995): Anti-inflammatory potency and gas-
`trointestinal toxicity of a new compound, NO-naproxen. Pharmacol
`Res 31:61–65.
`Davies NM, Roseth AG, Appleyard CB, McKnight W, Del Soldato P,
`Calignano A, Cirino G, Wallace JL (1996): NO-naproxen vs.
`naproxen: Ulcerogenic, analgesic and anti-inflammatory effects.
`Aliment Pharmacol Ther 11: 69–79.
`Douthwaite AH, Lintott SAM (1938): Gastroscopic observation of the
`effect of aspirin and certain other substances on the stomach. Lan-
`cet 2:1222–1225.
`Elliott SN, McKnight W, Cirino G, Wallace JL (1995): A nitric oxide-
`releasing nonsteroidal anti-inflammatory drug accelerates gastric
`ulcer healing in the rat. Gastroenterology 109:524–530.
`Fujihara CK, Malheiros DMAC, Antunes E, Donato JL, Santos MM,
`Sena CR, De Nucci G, Zatz R (1995): Nitroflurbiprofen, a novel
`nonsteroidal anti-inflammatory drug, limits glomerular injury in the
`remnant model (Abstract). J Am Soc Nephrol 6:1012.
`Garner A (1992): Adaptation in the pharmaceutical industry, with par-
`ticular reference to gastrointestinal drugs and diseases. Scand J
`Gastroenterol 27(193):83–89.
`
`Page 5 of 6
`
`Patent Owner Ex. 2063
`Mylan v. Pozen
`IPR2017-01995
`
`

`

`NO-RELEASING NSAIDS
`
`149
`
`Graham DY (1990): The relationship between nonsteroidal anti-inflam-
`matory drug use and peptic ulcer disease. Gastroenterol Clin North
`Am 19:171–182.
`JECFA (1996): Toxicological evaluation of certain food additives and
`contaminants. 44th Meeting of the Joint FAO/WHO Expert Com-
`mittee on Food Additives, International Programme on Chemical
`Safety, World Health Organization.
`Kubes P, Suzuki M, Granger DN (1991): Nitric oxide: An endog-
`enous modulator of leukocyte adhesion. Proc Natl Acad Sci USA
`88:4651–4655.
`Lopez-Belmonte J, Whittle BJR, Moncada S (1993): The actions of
`nitric oxide donors in the prevention or induction of injury to the
`rat gastric mucosa. Br J Pharmacol 108:73–78.
`MacNaughton WK, Cirino G, Wallace JL (1989): Endothelium-derived
`relaxing factor (nitric oxide) has protective actions in the stomach.
`Life Sci 45:1869–1876.
`Meade TW, Roderick PJ, Brennan PJ, Wilkes HC, Kelleher CC
`(1992): Extracranial bleeding and other symptoms due to low
`dose aspirin and low intensity oral anticoagulation. Thromb
`Haemost 68:1–6.
`Mitchell JA, Cirino G, Akarasereenont P, Wallace JL, Flower RJ, Vane
`JR (1994): Flurbinitroxybutylester: A novel anti-inflammatory drug
`devoid of ulcerogenic activity, inhibits cyclo-oxygenase-1 and cyclo-
`oxgenase-2. Can J Physiol Pharmacol 72:270.
`Moncada S, Palmer RMJ, Higgs EA (1991): Nitric oxide: Physiology,
`pathophysiology, and pharmacology. Pharmacol Rev 43:109–142.
`Patrono C (1994): Aspirin as an antiplatelet drug. N Engl J Med
`330:1287–1294.
`Reuter BK, Cirino G, Wallace JL (1994): Markedly reduced intestinal
`toxicity of a diclofenac derivative. Life Sci 55:PL1–PL8.
`Reuter BK, Davies NM, Wallace JL (1997): Nonsteroidal anti-inflam-
`matory drug-enteropathy in rats: Role of permeability, bacteria and
`enterohepatic circulation. Gastroenterology 112:109–117.
`The SALT Collaborative Group (1991): Swedish Aspirin Low-dose Trial
`(SALT) of 75 mg aspirin as secondary prophylaxis after cerebrovas-
`cular ischaemic events. Lancet 338:1345–1349.
`Segasothy M, Samad SA, Zulfigar A, Bennett WM (1994): Chronic
`renal disease and papillary necrosis associated with the long-term
`use of nonsteroidal anti-inflammatory drugs as the sole or predomi-
`nant analgesic. Am J Kidney Dis 24:17–24.
`Soll AH, Weinstein WM, Kurata J, McCarthy D (1991): Nonsteroidal
`anti-inflammatory drugs and peptic ulcer disease. Ann Intern Med
`114:307–319.
`
`Vane JR (1971): Inhibition of prostaglandin synthesis as a mechanism
`of action for aspirin-like drugs. Nat New Biol 231:232–235.
`Wallace JL (1993): Gastric ulceration: Critical events at the neutro-
`phil-endothelium interface. Can J Physiol Pharmacol 71:98–102.
`Wallace JL, Tigley AW (1995): New insights into prostaglandins and
`mucosal defence. Aliment Pharmacol Ther 9:227–235.
`Wallace JL, Keenan CM, Granger DN (1990): Gastric ulceration in-
`duced by nonsteroidal anti-inflammatory drugs is a neutrophil-de-
`pendent process. Am J Physiol 259:G462–G467.
`Wallace JL, Arfors K-E, McKnight GW (1991): A monoclonal anti-
`body against the CD18 leukocyte adhesion molecule prevents in-
`domethacin-induced gastric damage in the rabbit. Gastroenterology
`100:878–883.
`Wallace JL, McKnight W, Miyasaka M, Tamatani T, Paulson J, Ander-
`son DC, Granger DN, Kubes P (1993): Role of endothelial adhesion
`molecules in NSAID-induced gastric mucosal injury. Am J Physiol
`265:G993–G998.
`Wallace JL, Reuter B, Cicala C, McKnight W, Grisham MB, Cirino
`G (1994a): Novel nonsteriodal anti-inflammatory drug derivatives
`with markedly reduced ulcerogenic properties in the rat. Gastro-
`enterology 107:173–179.
`Wallace JL, Reuter B, Cicala C, McKnight W, Grisham MB, Cirino G
`(1994b): Adiclofenac derivative without ulcerogenic properties. Eur
`J Pharmacol 257:249–255.
`Wallace JL, Reuter BK, Cirino G (1994c): Nitric oxide releasing
`NSAIDs: A novel approach for reducing gastropathy. J Gastroenterol
`Hepatol 9:S40–S44.
`Wallace JL, McKnight W, Del Soldato P, Cirino G (1995a): Anti-throm-
`botic properties of a nitric oxide-releasing, gastric sparing aspirin
`derivative. J Clin Invest 96:2711–2718.
`Wallace JL, Pittman QJ, Cirino G (1995b): Nitric oxide-releasing
`NSAIDs: A novel class of GI-sparing anti-inflammatory drugs.
`Agents Actions 46:S121–S129.
`Wallace JL, McKnight W, Wilson TL, Del Soldato P, Cirino G (1997a):
`Reduction of shock-induced gastric damage by a nitric oxide-releasing
`aspirin derivative: Role of neutrophils. Am J Physiol 273:G1246–1251.
`Wallace JL, McKnight W, Wilson TL, Cirino G, Del Soldato P, Davies
`NM (1997b): Nitric oxide releasing aspirin derivative (NCX 4016):
`A gastric-sparing anti-thrombotic drug (Abstract). Gastroenterol-
`ogy 112:A325.
`Whittle BJR (1993): Neuronal and endothelium-derived mediators in
`the modulation of the gastric microcirculation: Integrity in the bal-
`ance. Br J Pharmacol 110:3–17.
`
`Page 6 of 6
`
`Patent Owner Ex. 2063
`Mylan v. Pozen
`IPR2017-01995
`
`

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