throbber
L
`E
`C
`T
`U
`R
`E
`From PGF2a-Isopropyl Ester to Latanoprost:
`A Review of the Development of Xalatan
`The Proctor Lecture
`
`Johan Wilhelm Stjernschantz
`
`T he research that led to the concept of using prostaglandins
`
`for reduction of intraocular pressure (IOP) has been dis-
`cussed by Bito and goes back to the early 1980s, when it was
`shown that PGF2a effectively reduces IOP in monkeys.1 Be-
`cause the IOP-reducing effect in primates was found to be
`profound and of long duration, it was of obvious interest to
`investigate whether prostaglandins could be developed into
`drugs for glaucoma treatment. A fruitful collaboration between
`Columbia University (New York, NY) and Pharmacia (Uppsala,
`Sweden), a pharmaceutical company, was initiated. As a result
`of the collaboration, a new glaucoma drug Xalatan was devel-
`oped. The purpose of this article is to present a review of the
`research that lead to the identification of latanoprost, and the
`development of Xalatan. Some relevant recent and previously
`unpublished data have been included as well. The experimen-
`tal protocols of all animal studies performed complied with the
`tenets of the ARVO Statement on the Use of Animals in Oph-
`thalmic and Vision Research, and all protocols were submitted
`for review and approval to the local Ethics Committee for
`Animal Experimentation. Protocols for clinical studies were
`submitted to the appropriate Ethics Committee/Internal Re-
`view Board and the Declaration of Helsinki (1964) with subse-
`quent revisions was adopted. Details concerning the experi-
`mental procedures of the previously unpublished results are
`given in the figure and table texts.
`
`PGF2a-Isopropyl Ester as a Prototype
`Prostaglandin Drug for Glaucoma Treatment
`The first approach to render prostaglandins suitable for glau-
`coma treatment was esterification of the carboxylic acid to
`improve the bioavailability2 and reduce the side effects. Several
`esters of PGF2a were synthesized and tested for IOP-reducing
`effect and side effects.3 One of the best of these prodrugs of
`PGF2a was the isopropyl ester (IE), but many other carboxylic
`acid esters, and di-, tri-, and tetra-esters and lactones of PGF2a
`were prepared and tested in addition (Bito LZ, Resul B, unpub-
`lished results, 1985). Similar experiments were also performed
`by researchers at Allergan Pharmaceuticals (Irvine, CA).4 Other
`companies (e.g., Ueno Fine Chemicals Industry, Osaka, Japan,
`and Alcon Laboratories, Fort Worth, TX) subsequently adopted
`the isopropyl ester prodrug concept for their respective pros-
`
`From the Department of Neuroscience, Unit of Pharmacology,
`Uppsala University, Uppsala, Sweden.
`Submitted for publication July 17, 2000; accepted September 8,
`2000.
`Commercial relationships policy: F, I, P. (The author is a former
`employee of Pharmacia and is a recipient of a research grant from
`Pharmacia, but has no commercial interest in Xalatan.)
`Corresponding author: Johan Stjernschantz, Department of Neu-
`roscience, Unit of Pharmacology, Box 593, Uppsala University Biomed-
`ical Center, S-751 24 Uppsala, Sweden.
`johan.stjernschantz@neuro.uu.se
`
`1134
`
`taglandin analogues (isopropyl unoprostone and travoprost,
`respectively).
`PGF2a-IE is a very efficacious and potent IOP-reducing agent
`in cats, dogs, and monkeys.2,5–7 Comparable reductions of IOP
`with PGF2a tromethamine salt requires a 10 to 100 times
`higher dose in monkeys.2,8 –10 Thus, esterification of PGF2a
`with isopropanol
`increased the bioavailability substantially.
`Overall, PGF2a-IE was found to be a very good IOP-reducing
`agent in many species except rabbits, in which a pressure
`increase frequently is induced by a breakdown of the blood–
`aqueous barrier. It is interesting that whereas cats exhibited
`distinct signs of ocular pain and discomfort (e.g., closure of the
`lids and lacrimation) unanesthetized monkeys usually did not
`(Stjernschantz J, unpublished results, 1988). In addition, in cats
`and dogs PGF2a and its esters are potent miotics.11
`In the first clinical trial with PGF2a-IE, which had the char-
`acter of a pilot test, no or a minimal IOP-reducing effect was
`observed in patients with glaucoma, probably because difficult
`cases were selected, refractory to all medical treatment (un-
`published results, Pharmacia). Despite these discouraging re-
`sults Villumsen and Alm,12 in cooperation with Pharmacia,
`started a systematic investigation to determine the IOP-reduc-
`ing effect and side effects of PGF2a-IE and found that the drug
`indeed effectively reduced IOP in a dose-dependent manner in
`healthy volunteers. However, the IOP-reducing effect was ac-
`companied by conjunctival hyperemia and ocular irritation
`similar
`to the side effects
`seen in studies with the
`tromethamine salt of PGF2a.13,14 The highest dose (10 mg) of
`PGF2a-IE caused pain and photophobia in all individuals.12 A
`dose of 0.5 mg twice daily was chosen for further studies in
`patients, and this dose, as well as a dose of 1 mg twice daily,
`was found to reduce IOP significantly, alone and in combina-
`tion with timolol.15–18
`However, many patients reported side effects such as for-
`eign-body sensation and conjunctival hyperemia, and it be-
`came questionable whether PGF2a-IE would offer any advan-
`tage over the already-established glaucoma medications. A
`particular problem was the irritative response to the drug. In
`animal experiments, it has been shown that PGF2a-IE induces
`albumin leakage in the iris and the ciliary body of monkeys at
`a dose of 1 mg,19 and it is possible that the 10 times higher dose
`previously used in healthy individuals12 induces edema in the
`anterior uvea that causes pain and photophobia.
`
`Effect of PGF2a-IE on the Uveoscleral Outflow
`Mode of Action
`The first evidence that PGF2a and its isopropyl ester reduces
`IOP through a mechanism based on increased uveoscleral
`outflow came from the studies by Crawford and Kaufman20
`and Nilsson et al.6 Both research groups independently of each
`other found evidence for increased uveoscleral outflow of
`aqueous humor in monkeys treated with PGF2a tromethamine
`salt or PGF2a-IE and no or minimal effect on the conventional
`
`Investigative Ophthalmology & Visual Science, May 2001, Vol. 42, No. 6
`Copyright © Association for Research in Vision and Ophthalmology
`
`Downloaded From: http://iovs.arvojournals.org/pdfaccess.ashx?url=/data/journals/iovs/933221/ on 05/11/2017
`
`Micro Labs Exhibit 1010
`
`

`

`IOVS, May 2001, Vol. 42, No. 6
`
`The Proctor Lecture
`
`1135
`
`TABLE 1. Effects of PGF2a-IE, PGF2b-IE, and 11-epi-PGF2a-IE on IOP, Pupil Diameter, and Nociception in Cats and on IOP in Monkeys
`
`Cat
`
`Monkey
`
`Prostaglandin
`Analogue
`
`Dose
`(mg)
`
`Reduction in IOP
`(mm Hg)
`
`Reduction in Pupil
`Diameter (mm)
`
`PGF2a-IE
`PGF2b-IE
`11-epi-PGF2a-IE
`
`1.0
`1.0
`1.0
`
`27.5 6 1.3‡
`28.3 6 1.5‡
`0.2 6 0.5
`
`29.3 6 0.4†
`0.0 6 0.0
`27.0 6 0.7†
`
`Irritation*
`
`2.7 6 0.2†
`1.5§
`1.0§
`
`Dose
`(mg)
`
`Reduction in IOP
`(mm Hg)
`
`FP Receptor EC50 Value
`(moles/l)
`
`3.0
`1.5
`2.4
`
`22.5 6 0.3†
`21.1 6 0.8
`22.0 6 0.9
`
`1.0 3 1028
`5.6 3 1026
`3.3 3 1028
`
`The values are based on the maximum difference between the experimental and contralateral control eyes (mean 6 SEM; n 5 5–6). The FP
`receptor EC50 values are based on results of in vitro tests performed on cat irides with the corresponding prostaglandin acids. Partly reproduced,
`with permission, from Resul et al., Surv Ophthalmol. 1997;41(suppl):S47–S52.
`* Arbitrary scale of 0 to 3 (see Fig. 3).
`† P , 0.001.
`‡ P , 0.005.
`§ Estimated mean value.
`
`outflow.6,20,21 Indirect evidence for a similar mechanism in
`humans was also obtained in two separate studies: PGF2a-IE
`was not found to have any effect on aqueous humor produc-
`tion or outflow facility.12,22 Thus, the most reasonable expla-
`nation for the IOP reduction was increased uveoscleral out-
`flow, although it could not be excluded that the drug may have
`reduced the episcleral venous pressure, too. It is important to
`note that in neither of the two clinical studies was any evi-
`dence found of a significant effect of PGF2a-IE on the blood–
`aqueous barrier.12,22
`
`Rationale of Receptor Selectivity for Elimination
`of Side Effects
`The preclinical and clinical studies with PGF2a-IE demon-
`strated that prostaglandins of the F type could be useful in the
`treatment of glaucoma, but it was not realistic to develop
`PGF2a-IE into a glaucoma drug for broader use, because of the
`side effects. Patients with severe disease could have endured
`treatment with the drug for some time. The question thus arose
`of whether it would be possible to separate the IOP-reducing
`effect from the side effects—primarily the irritative and hyper-
`emic effects— by changing the receptor profile of PGF2a
`through chemical modification. It should be recalled in this
`context that the classification of the prostanoid receptors in
`the mid 1980s was somewhat ambiguous, based on conven-
`tional pharmacologic experiments only.23–25 However, early
`experiments that we had performed with two epimers of
`PGF2a-IE, namely PGF2b-IE and 11-epi-PGF2a-IE indicated that
`the miotic and IOP responses to PGF2a-IE could be distinctly
`separated by these two epimers in cats (Table 1). PGF2b-IE
`reduced IOP with no miotic effect, whereas 11-epi-PGF2a-IE
`was a potent miotic with little effect on IOP. Furthermore, the
`epimers also differed from PGF2a-IE with respect to the noci-
`ceptive response (Table 1) and the hyperemic response,
`PGF2b-IE, being a much stronger vasodilator
`than both
`PGF2a-IE and 11-epi-PGF2a-IE (unpublished results; Pharmacia).
`Thus, it appeared possible to separate the different ocular
`responses from each other, at least partly, and there was also
`an indication that the FP prostanoid receptor, which mediates
`miosis in cats, may be involved at least partly in IOP reduction
`in monkeys.26 (Table 1).
`A critical aspect in the success of the screening work was
`the selection of appropriate animal models that would allow
`extrapolation of the results to the human eye. The cat eye
`seemed very unspecific, in that IOP reduction was brought
`about by widely different prostaglandin analogues (e.g., PGF2a,
`PGF2b, PGE2, PGA2, PGB2, and PGD2). Therefore, we regarded
`the cat eye as somewhat unpredictable with respect to the IOP
`effect in humans and used the cat eye primarily for studying
`
`the nociceptive and miotic effects, whereas conscious cyno-
`molgus monkeys were used to study the IOP-reducing effect.
`However, because young healthy monkeys usually have low
`IOP, often around 11 to 15 mm Hg, the test model was not
`ideal but was good enough to confirm whether an analogue
`had an IOP-reducing effect. The hyperemic effect was studied
`in albino rabbits, but there were no attempts to study anything
`else in the rabbit, because the rabbit eye is known to react very
`atypically to prostaglandins.27–29 Thus, the selection of ade-
`quate animal models was of paramount importance for the
`success of the project.
`
`Structure–Activity Approach and Identification of
`Phenyl-Substituted Prostaglandin Analogues
`The first approach to modifying PGF2a included various alter-
`ations of the carboxylic acid end of the molecule. The alter-
`ations comprised, for example, the alcohol and simple esters
`but generally did not result in any clear-cut improvement of the
`pharmacologic profile of PGF2a. The second approach was to
`change the stereochemistry, and the functional groups in the
`cyclopentane ring. Although this yielded some interesting an-
`alogues that offered certain advantages over PGF2a, such as
`11-epi-PGF2a,26 modifications of the cyclopentane ring re-
`sulted in no real breakthrough. The third approach was to alter
`parts of the vchain (e.g., the double bond between carbons 13
`and 14 and the 15-hydroxyl group) and to substitute part of the
`chain. However, it was well known that the 15-hydroxyl group
`is essential for biologic activity of prostaglandins, and dehydro-
`genation of the hydroxyl group results in marked loss of bio-
`logic activity.30 –31 Thus, the approach taken by the research-
`ers of Ueno Fine Chemicals Industry (Osaka, Japan) to reduce
`the side effects of PGF2a by preparing the 13,14-dihydro-15-
`keto metabolite, or modifications of this metabolite (e.g., iso-
`propyl unoprostone) renders molecules with significantly re-
`duced potency.32
`Among a group of different v-chain–modified prostaglan-
`din analogues, we quite unexpectedly noted that 17-phenyl-
`18,19,20-trinor-PGF2a-IE induced marked miosis in the cat
`without concomitant irritation, which almost all other pros-
`taglandin analogues had induced. Although there was no
`significant effect of the analogue on IOP in cats, conceptu-
`ally, the combination of marked miosis with absence of
`nociception demonstrated that it was possible to eliminate
`the nociceptive effect without losing pharmacologic activ-
`ity. In contrast to cats,33 monkeys responded to the com-
`pound with satisfactory IOP reduction.34 –35 The compound,
`which can be regarded as the breakthrough, was assigned
`the code name PhDH100A and became the lead compound
`of the group of v-chain ring-substituted prostaglandin ana-
`
`Downloaded From: http://iovs.arvojournals.org/pdfaccess.ashx?url=/data/journals/iovs/933221/ on 05/11/2017
`
`Micro Labs Exhibit 1010-2
`
`

`

`1136
`
`Stjernschantz
`
`IOVS, May 2001, Vol. 42, No. 6
`
`Influence of v-Chain Length on
`Pharmacologic Profile
`This aspect was studied in detail by attaching a terminal phenyl
`ring to carbons 15-24 (Fig. 1). The analogues were studied in
`cats with emphasis on the miotic and nociceptive responses.
`Of note, attaching the aromatic ring structure to carbon 17 of
`the prostaglandin skeleton yielded an optimal compound, in
`that the FP receptor function, as evident from the miotic
`response, was not compromised, whereas the nociceptive re-
`sponse was completely abolished34 (Table 2). In bizarre con-
`trast, 16-phenyl-17,18,19,20-tetranor-PGF2a-IE with one addi-
`tional carbon atom removed from the v chain caused
`significant irritation, albeit less than PGF2a-IE. Elongation of the
`v chain beyond carbon 17 with a terminal phenyl ring at-
`tached, reduced the biologic activity34 (Table 2). However, it is
`noteworthy that most analogues with a terminal ring structure
`exhibited considerably less nociceptive effect than PGF2a-IE.
`Substitution of carbon 17 with oxygen afforded a compound
`(16-phenoxy-17,18,19,20-tetranor-PGF2a-IE) with similar phar-
`macologic profile to that of 17-phenyl-18,19,20-trinor-PGF2a-IE
`(unpublished results; Pharmacia).
`
`Influence of Different Ring Structures on the
`Pharmacologic Profile
`A large number of compounds with different ring structures
`from cyclopropyl to cycloheptyl and aromatic ring structures,
`such as phenyl, thiophen, and furyl, attached to carbon 17 (Fig.
`1) were prepared and tested. Overall, these analogues exhib-
`ited an improved side-effect profile compared with PGF2a-IE,
`albeit with somewhat different pharmacologic activity. Thus it
`appears that many different terminal ring structures attached to
`carbon 17 in the v chain reduce the side effects of PGF2a-IE.
`
`Influence of Substituents in the Ring Structure on
`the Pharmacologic Profile
`Compounds with various substitutions in the phenyl ring (Fig.
`1) were also prepared and tested for pharmacologic activ-
`ity.26,34 Introduction of a methyl group into the ortho (2) or
`meta (3) position in the phenyl ring did not appreciably
`change the miotic activity of 17-phenyl-18,19,20-trinor-PGF2a-
`IE, whereas introduction of the methyl group into the para (4)
`position dramatically reduced the activity.26,34 Obviously, the
`methyl group in the para position induces a steric hindrance in
`the receptor ligand interaction. Introduction of an electron-
`attracting trifluormethyl group into the ortho or para position
`in the phenyl ring reduced the activity of the compound,
`
`FIGURE 1.
`Series of ring-substituted analogues of PGF2a-isopropyl es-
`ter tested for effects in the eye to determine the importance of v-chain
`length, ring structures, and substituents in the phenyl ring.
`
`logues in the screening for an optimal prostaglandin ana-
`logue for glaucoma treatment.
`
`Importance of v-Chain Length, Ring Structures,
`and Substituents
`The identification of 17-phenyl-18,19,20-trinor-PGF2a-IE lead to
`medicinal chemistry experiments that were of obvious interest
`to perform to understand the critical elements in the molecules
`for attaining efficacy and selectivity in the eye. Three aspects
`seemed particularly relevant to study: (1) the influence of the
`v-chain length, (2) the influence of different ring structures,
`and (3) the influence of substituents in the ring structure on
`the pharmacologic profile of the new analogues.
`
`TABLE 2. Effect of Phenyl-Substituted PGF2a-IE Analogues with Different v-Chain Lengths on IOP, Pupil Diameter,
`Nociception, and Conjunctival Hyperemia
`
`Cat
`
`Prostaglandin Analogue
`
`16-Phenyl-17,18,19,20-tetranor-PGF2a-IE
`17-Phenyl-18,19,20-trinor-PGF2a-IE
`18-Phenyl-19,20-dinor-PGF2a-IE
`19-Phenyl-20-nor-PGF2a-IE
`
`Monkey
`Reduction in IOP
`(mm Hg)
`
`Reduction in Pupil
`Diameter
`(mm)
`
`23.9 6 0.4†
`23.9 6 0.4†
`20.6 6 0.2§
`20.6 6 0.2§
`
`21.0 6 0.0†
`29.7 6 0.3†
`24.3 6 0.6†
`22.5 6 0.6‡
`
`Irritation*
`(0–3)
`
`2.2 6 0.3†
`0.0 6 0.0
`0.7 6 0.1†
`1.3 6 0.2‡
`
`Rabbit
`Hyperemia*
`(1–5)
`
`ND
`1.8 6 0.3‡
`0.3 6 0.7
`0.6 6 0.2§
`
`The values are based on the maximum difference between the experimental and contralateral control eyes. The dose was 3 mg in monkeys,
`1 mg in cats, and 0.5 mg in rabbits (mean 6 SEM; n 5 5–6; statistical significances determined by paired t-test). ND, not determined.
`* Arbitrary scale.
`† P , 0.001.
`‡ P , 0.01.
`§ P , 0.05.
`
`Downloaded From: http://iovs.arvojournals.org/pdfaccess.ashx?url=/data/journals/iovs/933221/ on 05/11/2017
`
`Micro Labs Exhibit 1010-3
`
`

`

`IOVS, May 2001, Vol. 42, No. 6
`
`The Proctor Lecture
`
`1137
`
`whereas introduction of the group into the meta position only
`slightly reduced the activity.26,34 Substituting fluorine for the
`trifluormethyl in the ortho, meta, orpara position afforded
`compounds with marked miotic effect and no or very little
`irritant effect in the cat eye, thus indicating that the trifluor-
`methyl group may also partly change the pharmacologic activ-
`ity through a steric effect.26,34 Introduction of an electron-
`donating methoxy group into the ortho or para position
`markedly reduced miotic activity, whereas introduction of the
`group into the meta position only slightly reduced miotic
`activity.26,34 The 16-(4-methoxy)-phenyl-17,18,19,20-tetranor
`PGF2a-IE analogue had virtually no irritant effect in the cat eye
`in contrast to 16-phenyl-17,18,19,20-tetranor-PGF2a-IE (unpub-
`lished results, Pharmacia). Thus, it appears that the para posi-
`tion, and to some extent the ortho position, in the phenyl ring
`are sensitive to steric hindrance, whereas the meta position is
`much less vulnerable. However, in the ortho position electro-
`chemical forces may be important, at least in part, because an
`electron-attracting trifluormethyl group reduces the activity in
`contrast to a neutral methyl group.
`Overall, the structure–activity studies indicated that the ring
`structure on the v chain is of paramount importance for
`reducing the side effects of PGF2a-IE, and furthermore that a
`large number of modifications of the ring structure are possi-
`ble, still affording useful compounds in the eye.26,34 –36
`Saturation of the 13,14-trans double bond of 17-phenyl-
`18,19,20-trinor-PGF2a-IE was found to further improve the re-
`ceptor profile somewhat, and 13,14-dihydro prostaglandin an-
`alogues in addition exhibited improved chemical stability. The
`13,14-dihydro-15R,S-17-phenyl-18,19,20-trinor-PGF2a-IE
`ana-
`logue was selected as the new candidate drug, and the com-
`pound was given the code name PhXA34. Because the 15R
`epimer is more potent than the 15S epimer, with time the 15R
`epimer (PhXA41) became the final candidate drug. It was given
`the generic name latanoprost and is the active principle in
`Xalatan. The chemical structures of PGF2a-IE, 17-phenyl-
`18,19,20-trinor-PGF2a-IE, PhXA34, and latanoprost (PhXA41)
`are presented in Figure 2.
`
`Latanoprost
`
`As is obvious from the structure–activity studies, the reason for
`the good therapeutic index of latanoprost in the eye is its
`pharmacologic receptor profile. It can be seen in Table 3 that
`latanoprost acid is a much more selective FP prostanoid recep-
`tor agonist than PGF2a. In practical terms it is even more
`selective than 17-phenyl-18,19,20-trinor-PGF2a because it spills
`less over on the EP1 and TP receptors (Table 3). It is also
`apparent that latanoprost acid is a full agonist on the FP
`receptor, and full or near full agonist on the EP1 and EP3
`receptors, but has no, or only weak effect on prostanoid
`receptors EP2, DP, IP, and TP (Table 3). In comparison 17-
`phenyl-18,19,20-trinor-PGF2a, with the 13,14 double bond in-
`tact, is a full agonist on the FP and EP1 receptors, and a partial
`agonist on the TP receptor, but has no, or only weak effect on
`the other receptors (Table 3). Thus, increasing the flexibility of
`the v chain by saturating the 13,14 double bond has relatively
`little effect on the interaction with the FP receptor, but re-
`duces the potency on the EP1 and TP receptors, and increases
`the capacity to stimulate the EP3 receptor, albeit only at very
`high concentrations.
`Latanoprost has virtually no IOP-reducing effect in cats or
`rabbits, but induces a moderate IOP reduction in conscious
`normotensive monkeys as measured 3 to 6 hours after topical
`application. During continuous treatment with 3 mg once daily
`for 5 days the IOP reduction lasted around the clock (unpub-
`lished results; Pharmacia). In ocular hypertensive monkeys a
`good IOP-reducing effect of latanoprost has also been seen.37
`
`FIGURE 2. Chemical structures of PGF2a-IE, 17-phenyl-18,19,20-trinor-
`PGF2a-IE, 13,14-dihydro-15R, S-17-phenyl-18,19,20-trinor-PGF2a-IE (PhXA34),
`and 13,14-dihydro-15R-17-phenyl-18,19,20-trinor-PGF2a-IE (latanoprost).
`
`The absence of effects in cats and rabbits may be due to several
`factors—for example, different anatomy of the ciliary muscle
`and aqueous humor outflow pathways compared with pri-
`mates, and the absence of expression or different coupling of
`FP receptors in the ciliary muscle. Of note, recently it was
`demonstrated that the prostanoid receptor EP1 seems to medi-
`ate the IOP reduction of PGF2a in the cat.38
`
`Mode of Action
`Thorough pharmacodynamic studies in cannulated monkey
`eyes were performed to investigate the mode of action of
`latanoprost, because the mechanism could differ from that of
`PGF2a-IE due to the different receptor profiles of the com-
`pounds. Cynomolgus monkeys were treated for 5 days topi-
`cally on one eye with 3 mg latanoprost daily; the other eye
`received vehicle only and served as the control. A detailed
`description of the pharmacodynamic method has been pre-
`sented.39 The results showed that latanoprost had a statistically
`significant effect on the uveoscleral outflow, which increased
`by approximately 60% compared with the contralateral control
`eye.39,40 Neither the trabecular outflow of aqueous humor, nor
`the total outflow facility changed during latanoprost treat-
`ment.39,40
`The reason for the increase in flux of aqueous humor
`through the ciliary muscle during prostaglandin treatment has
`been studied in detail at the cellular level by Lindsey et al.41– 47
`PGF2a seems to increase the expression of c-Fos in human
`ciliary muscle cells, which in turn may increase the expression
`of matrix metalloproteinases (MMPs)—for example, MMP-1,
`MMP-2, MMP-9 and MMP-10 —as well as their precursors,42,43
`thereby perturbing the balance in the turnover of extracellular
`
`Downloaded From: http://iovs.arvojournals.org/pdfaccess.ashx?url=/data/journals/iovs/933221/ on 05/11/2017
`
`Micro Labs Exhibit 1010-4
`
`

`

`1138
`
`Stjernschantz
`
`IOVS, May 2001, Vol. 42, No. 6
`
`TABLE 3. Potency Based on EC50 Values and Estimated Efficacy of PGF2a, 17-Phenyl-18,19,20-trinor-PGF2a (17-Phenyl-PGF2a), and Latanoprost
`Acid as Measured in Functional Receptor Assays24
`
`FP
`
`EP1
`
`EP2
`
`EP3
`
`IP/DP
`
`TP
`
`Prostaglandin
`Analogue
`
`Potency
`
`Efficacy
`
`Potency
`
`Efficacy
`
`Potency
`
`Efficacy
`
`Potency
`
`Efficacy Potency Efficacy
`
`Potency
`
`Efficacy
`
`1.2 3 1028
`PGF2a
`17-Phenyl-PGF2a 4.5 3 1029
`1.0 3 1028
`Latanoprost acid
`
`100
`100
`100
`
`3.2 3 1027
`6.5 3 1027
`5.0 3 1026
`
`100
`100
`100
`
`6.4 3 1026
`.1024
`.1024
`
`100
`0
`0
`
`1.6 3 1027
`.1024
`2.8 3 1025
`
`100
`0
`;80
`
`.1024
`.1024
`.1024
`
`0
`0
`0
`
`.1024
`3.4 3 1025
`.1024
`
`;20
`;50
`0
`
`The receptor assays were based on the following tissues: FP, cat iris sphincter; EP1, bovine iris sphincter in the presence of GR32191B; EP2,
`electrical stimulation of guinea pig ileum; EP3, electrical stimulation of guinea pig vas deferens; IP/DP, prevention of adenosine diphosphate-
`induced guinea pig platelet aggregation; and TP, guinea pig platelet aggregation. For estimation of efficacy, the compounds were compared with
`PGF2a (FP), PGE2 (EP1, EP2, and EP3), carbaprostacyclin (IP), BW245C (DP), and U-46619 (TP). Potency is expressed in moles per liter and efficacy
`as percentage.
`
`matrix components toward catabolism.44 – 46 PGF2a-IE was
`found to reduce collagens I, III, and IV in the ciliary muscle and
`adjacent sclera of the monkey after topical treatment with 2 mg
`twice daily for 5 days.47 Similar results were obtained by
`Ocklind48 who demonstrated a decrease in collagens I, III, and
`IV; laminin; fibronectin; and hyaluronan in human ciliary mus-
`cle cell cultures exposed to latanoprost acid in parallel with an
`increase in MMP-2 and MMP-3. She also found evidence for
`reduced collagens IV and VI levels in the ciliary muscle after 10
`days of topical treatment with 3 mg latanoprost daily in mon-
`keys.48 Furthermore, evidence for a change in the shape of
`ciliary muscle cells was also found after exposure to latano-
`prost acid in vitro, with alterations in the actin and vinculin
`localization in the cells.39 Thus, the results indicate that latano-
`prost may have complex effects on ciliary muscle, the net
`effect being increased percolation of aqueous humor through
`the tissue.
`
`Vascular Effects of Latanoprost
`Both the local and systemic vascular effects of latanoprost have
`been studied in detail. In the rabbit eye latanoprost induced no
`or minimal change in blood flow,19 in sharp contrast to PGF2a-
`IE, which induced marked increase in blood flow to the surface
`structures and the anterior uvea after topical application.49 The
`hyperemic effect of PGF2a-IE seems to be based on a release of
`nitric oxide (NO), and apparently the mechanism leading to
`NO release does not involve FP receptors.49 Of interest, sen-
`
`sory denervation by electrocoagulation of the ophthalmic
`nerve, almost completely abolished the hyperemic effect of
`PGF2a-IE in rabbits, implying that the effect is nerve-medi-
`ated.50 This fits well with the absence of nociceptive effect of
`FP prostanoid receptor agonists such as latanoprost. By using
`selective agonists we studied which of the prostanoid recep-
`tors mediate the nociceptive response to prostaglandins in the
`cat eye and found that the FP and EP2 receptors are of little or
`no importance (Fig. 3). Stimulation of the DP, IP, EP1, and EP3
`receptors, on the contrary, induced a nociceptive response in
`the cat eye (Fig. 3). Whether PGF2a-IE–induced conjunctival
`hyperemia in humans also involves sensory nerves is unknown,
`but it is quite possible.
`Latanoprost and PhXA34 tested at a dose of approximately
`four times the clinical dose of latanoprost in Xalatan had a
`negligible effect on the regional blood flow in the monkey eye
`after topical application.51 Neither was any effect seen on
`capillary permeability to albumin in the monkey eye.51 Intra-
`venous injection of latanoprost in escalating doses of up to 6
`mg/kg body weight had no statistically significant effect on the
`uveal or retinal blood flow in monkeys, although a tendency
`toward increased blood flow was observed (Table 4).19 In
`aphakic monkey eyes with intact posterior lens capsule, latano-
`prost induced no capillary leakage in the retina as studied with
`fluorescein angiography during 6 months of treatment, and
`similar results were also obtained during shorter treatment
`periods in pseudophakic patients.52 Thus, it appears that the
`
`FIGURE 3. Maximum nociceptive effect of selective prostanoid receptor agonists in the cat eye. The
`agonists were applied unilaterally at three different doses. The irritation was estimated by observing the
`animals for 6 hours after administration of the test substances. An arbitrary scale of 0 to 3 was used: 0, no
`signs of ocular irritation; 1, slight; 2, moderate; and 3, marked signs of irritation as evident from complete
`lid closure and lacrimation. The complete name of the EP3 agonist is 13,14-dihydro-D14-trans-15-deoxy-
`17-phenyl-18,19,20-trinor-PGF2a-IE, and the compound is not a full agonist on the EP3 receptor. Thus, the
`effect on the EP3 receptor may be underestimated. Mean 6 SEM; n 5 4 – 6 for each compound and dose;
`*P , 0.01, paired t-test. IE, isopropyl ester; ME, methyl ester.
`
`Downloaded From: http://iovs.arvojournals.org/pdfaccess.ashx?url=/data/journals/iovs/933221/ on 05/11/2017
`
`Micro Labs Exhibit 1010-5
`
`

`

`IOVS, May 2001, Vol. 42, No. 6
`
`The Proctor Lecture
`
`1139
`
`TABLE 4. Effect of Intravenous Injection of Escalating Doses of Latanoprost on the Blood Flow in the Monkey Eye and Vital Organs
`as Studied with Radioactively Labeled Microspheres19
`
`Baseline
`
`0.6 mg/kg
`
`2 mg/kg
`
`6 mg/kg
`
`Ocular tissues
`Retina
`Choroid
`Ciliary body
`Iris
`
`Other tissues
`Frontal brain
`Occipital brain
`Parietal brain
`Cerebellum
`Choroid plexus
`Myocardium right atrium
`Myocardium left atrium
`Myocardium right ventricle
`Myocardium left ventricle
`Liver
`Stomach
`Ileum
`Colon
`Kidney
`
`27 6 3
`570 6 95
`80 6 14
`12 6 2
`
`0.66 6 0.06
`1.64 6 0.34
`0.56 6 0.06
`0.54 6 0.07
`5.02 6 0.54
`0.40 6 0.09
`0.44 6 0.11
`1.35 6 0.18
`1.90 6 0.21
`0.26 6 0.05
`0.24 6 0.04
`0.82 6 0.14
`0.55 6 0.10
`5.80 6 0.58
`
`25 6 4
`574 6 92
`83 6 12
`13 6 2
`
`0.70 6 0.07
`1.73 6 0.36
`0.59 6 0.07
`0.50 6 0.05
`4.94 6 0.38
`0.42 6 0.06
`0.47 6 0.11
`1.58 6 0.18
`2.10 6 0.18
`0.26 6 0.05
`0.24 6 0.04
`0.89 6 0.14
`0.56 6 0.07
`5.53 6 0.45
`
`30 6 3
`670 6 119
`103 6 19
`12 6 2
`
`0.81 6 0.09
`2.00 6 0.50
`0.68 6 0.10
`0.55 6 0.05
`5.59 6 0.60
`0.73 6 0.20*
`0.60 6 0.17
`2.28 6 0.38
`2.79 6 0.46
`0.32 6 0.12
`0.21 6 0.03
`0.93 6 0.13
`0.55 6 0.11
`5.42 6 0.42
`
`40 6 4
`669 6 99
`105 6 16
`15 6 2
`
`1.09 6 0.16*
`1.69 6 0.29
`0.88 6 0.13*
`0.61 6 0.08
`6.06 6 0.70
`1.29 6 0.29*
`0.89 6 0.22*
`4.68 6 1.10*
`4.73 6 1.01*
`0.26 6 0.08
`0.40 6 0.10
`1.36 6 0.23*
`0.60 6 0.06
`5.93 6 0.42
`
`The blood flow in the eye is expressed per whole tissue and represents the right eye. Eye tissue data are in milligrams per minute; other tissue
`data are in grams per minute per gram tissue weight. (Mean 6 SEM; n 5 5–7.)
`* P , 0.05 compared with baseline (ANOVA; Tukey’s test).
`
`FP receptor, if expressed in the vasculature, plays no or only a
`limited role in the regulation of vessel tone and capillary
`permeability in the eye.
`The effects of latanoprost on the systemic circulation have
`been studied in monkeys after intravenous administration.
`With escalating doses of up to 6 mg/kg body weight an increase
`of the blood flow in parts of the brain as well as the heart was
`found, whereas very little effect was seen in other vital organs,
`such as the liver, gastrointestinal tract, and kidneys (Table 4).
`A tendency toward a slight increase in blood pressure was
`found. This effect seems to be based on increased cardiac
`output induced by high doses of the drug in anesthetized
`animals.19 It should be emphasized that the highest dose of 6
`mg/kg body weight is approximately 100 times the clinical
`dose of latanoprost in Xalatan (per body weight) applied top-
`ically on the eye.
`
`Effects of Latanoprost on the Respiratory System
`As PGF2a is a well-known constrictor of human bronchi,53–56
`the effect of latanoprost on pulmonary function in healthy
`volunteers and patients who have bronchial asthma was im-
`portant to investigate. No negative effects on pulmonary func-
`tion were observed in two specially designed clinical stud-
`ies.57,58 In monkeys, however, intravenous infusion of high
`doses of latanoprost was found to increase the intrathoracic
`inspiration– expiration pressure difference consistent with
`bronchoconstriction (unpublished results; Pharmacia). A study
`was therefore undertaken to determine the effects of latano-
`prost acid on human bronchioles in vitro. Both PGF2a and
`latanoprost acid contracted the smooth muscle of the bronchi-
`oles, as measured in small-vessel myographs with an EC50 value
`of approximately 1026 M. Latanoprost acid exerted about half
`the maximum effect of PGF2a.59 Both the contraction to PGF2a
`and latanoprost acid was completely abolished by a TP recep-
`tor antagonist (GR32191B). Thus, it appears that the effect is
`mediated primarily by TP receptors and not FP receptors. A
`concentration of 1027 M was necessary to elicit any response
`at all to latanoprost acid.59 This concentration exceeds the
`maximum concentration in

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