throbber
Trials@uspto.gov
`571-272-7822
`
`
`
`
`Paper 37
`Entered: July 19, 2017
`
`
`
`
`UNITED STATES PATENT AND TRADEMARK OFFICE
`____________
`
`BEFORE THE PATENT TRIAL AND APPEAL BOARD
`____________
`
`MYLAN PHARMACEUTICALS INC.
`and MYLAN LABORATORIES LIMITED,
`Petitioner,
`
`v.
`
`UCB PHARMA GMBH,
`Patent Owner.
`____________
`
`Case IPR2016-00517
`Patent 7,985,772 B2
`____________
`
`
`Before KRISTINA M. KALAN, ROBERT A. POLLOCK, and
`MICHELLE N. ANKENBRAND, Administrative Patent Judges.
`
`POLLOCK, Administrative Patent Judge.
`
`FINAL WRITTEN DECISION
`Finding Claims 1, 3, 4, and 6–8 Not Unpatentable
`35 U.S.C. § 318(a) and 37 C.F.R. § 42.73
`Dismissing as Moot Patent Owner’s Motion to Exclude
`37 C.F.R. § 42.64(c)
`Granting Joint Motion to Seal and Entering Default Protective Order
`37 C.F.R. § 42.54
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`
`I. INTRODUCTION
`Mylan Pharmaceuticals Inc. and Mylan Laboratories Limited
`(“Mylan” or “Petitioner”) filed a Corrected Petition requesting an inter
`partes review of claims 1, 3, 4, and 6–8 of U.S. Patent No. 7,985,772 B2
`(Ex. 1001, “the ’772 patent”). Paper 5 (“Pet.”). UCB Pharma GmbH,
`(“UCB” or “Patent Owner”) filed a Preliminary Response to the Petition.
`Paper 9 (“Prelim. Resp.”). We instituted an inter partes review of claims 1,
`3, 4, and 6–8 on the grounds of unpatentability alleged in the Petition. Paper
`12 (“Inst. Dec.”). After institution of trial, Patent Owner filed a Patent
`Owner Response (Paper 17, “PO Resp.”) and Petitioner filed a Reply (Paper
`21, “Reply”). An oral hearing was held on April 5, 2017. A transcript of the
`hearing has been entered into the record. Paper 36 (“Tr.”).
`The Board has jurisdiction under 35 U.S.C. § 6. In this Final Written
`Decision, issued pursuant to 35 U.S.C. § 318(a) and 37 C.F.R. § 42.73, we
`determine that Petitioner has not shown by a preponderance of the evidence
`that claims 1, 3, 4, and 6–8 of the ’772 patent are unpatentable under
`35 U.S.C. § 103.
`
`II. BACKGROUND
`
`A. Related Matters
`Patent Owner asserts that
`UCB and Pfizer Inc. (“Pfizer”), the exclusive licensees of the ‘772
`patent, have sued Mylan Pharmaceuticals Inc. for infringement of
`the ‘772 patent in the following actions: Pfizer, Inc. and UCB
`Pharma GMBH v. Mylan Pharmaceuticals, Inc., No. 1:15-cv-
`00079-GMS (D. Del.) and Pfizer Inc. and UCB Pharma GMBH
`v. Mylan Pharmaceuticals Inc., Case No. 1:15-cv-00013-IMK
`(N.D.W.Va.).
`Paper 8, 2; see Pet. 1–2 (noting that Pfizer is the NDA holder).
`
`
`
`2
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`
`The ’772 patent is also at issue in Pfizer, Inc. v. Sandoz, Inc., No. 1:13-
`cv-01110-GMS (D. Del.),1 and in the now-dismissed action, Pfizer, Inc. v.
`Dr. Reddy’s Laboratories, Ltd., No. 1:15-cv-01067 (GMS) (D. Del.). Paper
`8, 2.
`
`In addition to the case before us, we instituted inter partes review in
`the following matters involving patents with substantially the same
`specification as the ’772 patent at issue here:
`Case No. IPR2016-00512 (U.S. Patent No. 7,384,980 B2);
`Case No. IPR2016-00514 (U.S. Patent No. 7,855,230 B2); and
`Case No. IPR2016-00516 (U.S. Patent No. 8,338,478 B2).
`We also instituted inter partes review in IPR2016-00510 (U.S. Patent
`No. 6,858,650 B1), a matter involving another UCB patent generally
`directed, as are the above patents, to 3,3-diphenylpropylamine compounds.2
`Patent Owner updated its mandatory notices on February 16, 2017, to
`reflect that Case No. 1:15-cv-00079-GMS concluded with a general verdict
`in favor of Plaintiffs, and that UCB Pharma GmbH and Pfizer Inc., the
`exclusive licensee of the asserted patents, filed suit against Torrent
`Pharmaceuticals Limited and Torrent Pharma Incorporated for infringement
`
`
`1 Patent Owner provides, as Exhibit 2001, the District Court’s Memorandum
`finding that the defendants in that proceeding “failed to present a prima facie
`case that the asserted claims of the patents-in-suit are invalid as obvious.”
`Ex. 2001, 19. Although the district court reached this determination on a
`different record and applied different standards, the arguments and references
`applied overlap with those before us. See Ex. 2001. Accordingly, while we
`are not bound to these findings, we find the court’s analysis informative.
`2 Petitioners Alembic Pharmaceuticals Limited from IPR2016-01596, Torrent
`Pharmaceuticals Limited from IPR2016-01636, and Amerigen
`Pharmaceuticals Limited from IPR2016-01665 were joined as Petitioners to
`IPR2016-00510. IPR 2016-00510, Papers 24–26.
`
`
`
`3
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`of the asserted patents in Pfizer, Inc. and UCB Pharma GmbH v. Torrent
`Pharm. Ltd., No. 1:17-cv-00112-GMS (D. Del.). Paper 26, 2.
`
`B. The ’772 Patent
`The ’772 patent, entitled “Derivatives of 3,3-Diphenylpropylamines,”
`issued on July 26, 2011, with Claus Meese and Bengt Sparf as the listed co-
`inventors. Ex. 1001. The ’772 patent is generally directed to “derivatives of
`3,3-diphenylpropylamines, methods for their preparation, pharmaceutical
`compositions containing the novel compounds, and the use of the compounds
`for preparing drugs.” Id. at Abstract.
`The Specification discloses that “normal urinary bladder contractions
`are mediated mainly through cholinergic muscarinic receptor stimulation.”
`Id. at 1:23–24. Because the same muscarinic receptors appear to also
`mediate contractions of the overactive bladder and associated symptoms of
`urinary frequency, frequency urge, and urge incontinence, antimuscarinic
`drugs have been proposed for the treatment of bladder overactivity. Id. at
`1:25–30. “Among the antimuscarinic drugs available on the market,
`oxybutynin is currently regarded as the gold standard for pharmacological
`treatment of urge incontinence and other symptoms related to bladder
`overactivity” but its usefulness is limited by antimuscarinic side effects, most
`particularly, dry mouth. Id. at 1:31–34.
`“Tolterodine is a new, potent and competitive, muscarinic receptor
`antagonist intended for the treatment of urinary urge incontinence and
`[bladder wall muscle] hyperactivity. Preclinical pharmacological data show
`that tolterodine exhibits a favourable tissue selectivity in vivo for the urinary
`bladder over the effect on the salivation” as compared to oxybutynin. Id. at
`1:42–48.
`
`
`
`4
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`
`A major metabolite of tolterodine, the 5-hydroxymethyl derivative
`5-HMT (“5-HMT”), shows in vitro and in vivo pharmacological profiles
`almost identical to those of tolterodine. Id. at 1:55–59 (citing Nilvebrant et
`al., 1997, Eur. J. Pharmacol. 327 (1997), 195–207). “WO 94/11337 proposes
`[5-HMT] as a new drug for urge incontinence.” Id. at 1:63–64.
`The chemical structures of tolterodine and its active metabolite,
`5-HMT (indicated below by “5-HM”), are shown below:
`
`
`
`See, e.g., Pet. 19; Ex. 1010, 289; Ex. 1011, 530. As illustrated above,
`tolterodine has a single hydroxyl group at the 2-position carbon of the
`methylated phenolic ring, whereas 5-HMT bears a second hydroxyl moiety
`on the 5-position methyl group of that ring.
`
`C. Illustrative Claim
`Claim 1 recites:
`1. 3,3-Diphenylpropylamines of the general formula
`
`
`
`wherein:
`R1 is a hydrogen and R2 is C1-C6 alkylcarbonyl; or
`R1 is C1-C6 alkylcarbonyl and R2 is hydrogen;
`
`
`
`5
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`
`their salts with physiologically acceptable acids, their free
`bases and, when the 3,3-Diphenylpropylamines are in
`the form of optical isomers, the racemic mixture and the
`individual enantiomers.
`
`Claim 2 specifies that R1 is a hydrogen and R2 is C1-C6 alkylcarbonyl;
`claims 4, 6, and 7 recite methods of treating urinary incontinence using the
`compounds of claims 1 and 2; and claim 8 recites a pharmaceutical
`composition comprising those compounds and a pharmaceutically acceptable
`carrier.
`The compositions of claims 1 and 2 encompass fesoterodine fumarate
`(R-(+)-2-(3-(diisopropylamino-1-phenylpropyl)-4-hydroxymethl-
`phenylisobutyrate ester hydrogen fumarate) distributed by Pfizer Labs under
`the tradename Toviaz. See Pet. 6; PO Resp. 62; Ex. 1024, 8, 19.
`
`D. Instituted Grounds of Unpatentability
`We instituted inter partes review of claims 1, 3, 4, and 6–8 of the ’772
`patent on the following grounds:
`
`
`
`6
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`
`Basis
`§ 103
`
`§ 103
`
`Claims Challenged
`1, 3, 4, and 6–8
`
`1, 3, 4, and 6–8
`
`References
`Postlind,3 “Bundgaard
`publications,”4,5,6 Detrol
`Label,7 and Berge8
`Brynne,9 Bundgaard, and
`Johansson10
`In support of its challenges, Petitioner relies also on the Declaration of
`its technical expert Steven E. Patterson, Ph.D. (Ex. 1003) and the Declaration
`of DeForest McDuff, Ph.D. (Ex. 1033) with respect to lack of commercial
`success. Patent Owner relies on the Declaration of Dr. Hans Maag
`(Ex. 2021), the Declaration of William R. Roush, Ph.D. (Ex. 2022), the
`Declaration of Dr. Scott A. MacDiarmid (Ex. 2023), the Declaration of
`Leonard J. Chyall, Ph.D. (Ex. 2024), and the Declaration of Claus O. Meese,
`Ph.D. (Ex. 2025).
`
`
`3 Postlind et al., Tolterodine, A New Muscarinic Receptor Antagonist, is
`Metabolized by Cytochromes P450 2D6 and 3A in Human Liver Microsomes,
`26(4) DRUG METABOLISM & DISPOSITION 289–293 (1998). Ex. 1010
`(“Postlind”).
`4 In the Institution Decision, we interpreted Petitioner’s reference to
`“Bundgaard publications” as referring to Exhibits 1012 and 1020. Inst. Dec.
`6 n.3. We discuss those Exhibits individually in our analysis herein.
`5 Bundgaard, Design of Prodrugs, Elsevier (1985). Ex. 1012 (“Bundgaard”).
`6 WO 92/08459, published May 29, 1992. Ex. 1020 (“Bundgaard PCT”).
`7 Detrol™ (tolterodine tartrate tablets) prescribing information (1998).
`Ex. 1009 (“Detrol Label”).
`8 Berge et al., Pharmaceutical Salts, 66(1) J. PHARM. SCI. 1–19 (1977).
`Ex. 1013 (“Berge”).
`9 Brynne et al., Influence of CYP2D6 polymorphism on the pharmacokinetics
`and pharmacodynamics of tolterodine, 63(5) CLIN. PHARMACOL. &
`THERAPEUTICS 529–539 (1998). Ex. 1011 (“Brynne”).
`10 Johansson et al., WO 94/11337, published May 26, 1994. Ex. 1005
`(“Johansson”).
`
`
`
`7
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`E. Level of Ordinary Skill in the Art
`For the purpose of this decision, we accept Petitioner’s undisputed
`contention that:
`[a] person of ordinary skill in the art would have a Ph.D. in
`chemistry, medicinal chemistry, pharmacology, or a related field,
`and at least one year of industrial exposure to drug discovery,
`drug design, and synthesis. In lieu of an advanced degree, the
`individual may have additional years of industry experience,
`including, for example, in drug discovery, drug synthesis, and
`structure-activity work.
`Pet. 6 (citing Ex. 1003 ¶ 20); see Prelim. Resp. 9; PO Resp. 6.
`Based on our review of the ’772 patent, the types of problems and
`solutions described in the ’772 patent and cited prior art, we adopt
`Petitioner’s definition of a person of ordinary skill in the art at the time of the
`claimed invention. We note that the applied prior art also reflects the
`appropriate level of skill at the time of the claimed invention. See Okajima v.
`Bourdeau, 261 F.3d 1350, 1355 (Fed. Cir. 2001).
`
`III. ANALYSIS
`
`A. Claim Interpretation
`The Board interprets claim terms in an unexpired patent according to
`the broadest reasonable construction in light of the specification of the patent
`in which they appear. See Cuozzo Speed Techs., LLC v. Lee, 136 S. Ct. 2131,
`2144–46 (2016) (upholding the use of the broadest reasonable interpretation
`standard); 37 C.F.R. § 42.100(b). Under that standard, and absent any special
`definitions, we give claim terms their ordinary and customary meaning, as
`would be understood by one of ordinary skill in the art at the time of the
`invention. See In re Translogic Tech., Inc., 504 F.3d 1249, 1257 (Fed. Cir.
`2007). Only those terms which are in controversy need to be construed, and
`
`
`
`8
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`only to the extent necessary to resolve the controversy. See Vivid Techs., Inc.
`v. Am. Sci. & Eng’g, Inc., 200 F.3d 795, 803 (Fed. Cir. 1999).
`Petitioner submits that the “claims in the ʼ772 patent are presumed to
`take on their ordinary and customary meaning based on the broadest
`reasonable interpretation of the claim language.” Pet. 6. Patent Owner “does
`not dispute Petitioner’s position that no claim terms require construction,
`which the Board also accepted for purposes of institution.” PO Resp. 7
`(citing Pet. 6; Inst. Dec. 7). In our Decision on Institution, we determined
`that “for purposes of this Decision, none of the terms in the challenged claims
`require express construction at this time.” Inst. Dec. 7. As in the Decision
`on Institution, we apply the broadest reasonable interpretation standard here,
`and see no reason to construe explicitly any claim term.
`
`B. Principles of Law
`A claim is unpatentable under 35 U.S.C. § 103(a) if the differences
`between the subject matter sought to be patented and the prior art are such
`that the subject matter as a whole would have been obvious at the time the
`invention was made to a person having ordinary skill in the art to which said
`subject matter pertains. KSR Int’l Co. v. Teleflex Inc., 550 U.S. 398, 406
`(2007). The question of obviousness is resolved on the basis of underlying
`factual determinations including: (1) the scope and content of the prior art;
`(2) any differences between the claimed subject matter and the prior art;
`(3) the level of ordinary skill in the art; and (4) objective evidence of
`nonobviousness. Graham v. John Deere Co., 383 U.S. 1, 17–18 (1966). A
`decision on the ground of obviousness must include “articulated reasoning
`with some rational underpinning to support the legal conclusion of
`obviousness.” In re Kahn, 441 F.3d 977, 988 (Fed. Cir. 2006). The
`
`
`
`9
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`obviousness analysis “should be made explicit” and it “can be important to
`identify a reason that would have prompted a person of ordinary skill in the
`relevant field to combine the elements in the way the claimed new invention
`does.” KSR, 550 U.S. at 418. We analyze the asserted grounds of
`unpatentability in accordance with the above-stated principles.
`
`C. Overview of the Asserted References
`We begin our discussion with a brief summary of the references
`asserted.11
`Postlind (Ex. 1010)
`1.
`Postlind investigates the metabolism of tolterodine in human liver
`microsomes having varying P450 cytochrome activities. Ex. 1010, Abstract.
`Postlind illustrates the results of these studies in Figure 1, reproduced below.
`
`
`Figure 1 illustrates that “[m]etabolites are formed via two pathways:
`oxidation of the 5-methyl group to a 5-hydroxymethyl derivative (5-HM)
`
`
`11 Unless otherwise noted, we refer to the original page numbers in each
`reference, and not the page numbers Petitioner has added to the document.
`
`
`
`10
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`[i.e., 5-HMT]” by cytochrome P450 2D6 (“CYP2D6” or “2D6”) “and
`dealkylation of the nitrogen” by cytochrome P450 CYP3A4 (“CYP3A4”).
`Id. at 289; see also id. at 292 (concluding that the dealkylation reaction “is
`predominantly catalyzed by CYP3A4 in human liver microsomes.”)12
`Postlind notes that “[c]linical studies have demonstrated that
`individuals with reduced CYP2D6-mediated metabolism represent a high-
`risk group in the population with a propensity to develop adverse drug
`effects” and a “number of drugs [have been] identified as being affected by
`CYP2D6 polymorphism . . . . Id. at 292. Accordingly, “[t]he possibility of
`clinical drug interaction at the enzyme level [] exists, especially if tolterodine
`is administered at the same time as a compound that is preferentially
`metabolized by CYP2D6 or to individuals associated with the CYP2D6 poor
`metabolizer phenotype.” Id.
`Postlind further notes that CYP3A enzymes (e.g., CYP3A4) also have
`been associated with adverse drug interactions; “[h]owever, the large amount
`of CYP3A in the liver and the fact that tolterodine is predominantly
`eliminated via oxidation by CYP2D6 makes it less likely that clinically
`significant drug-drug interactions would occur with CYP3A substrates in
`individuals with the CYP2D6 extensive metabolizer phenotype.” Id.
`
`
`12 Petitioner’s technical expert, Dr. Patterson, emphasizes that 5-HMT is also
`N-dealkylated by CYP3A4. Ex. 1003 ¶ 111; see id. at ¶¶ 45–47 (citing
`Brynne et al., Pharmacokinetics and pharmacodynamics of tolterodine in
`man: a new drug for the treatment of urinary bladder overactivity, 35(7)
`INT’L J. CLIN. PHARMACOL. THERAP. 287–95 (1997) (Ex. 1007, “Brynne
`1997”); Ex. 1007, 291 Fig. 2.
`
`
`
`11
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`
`Brynne (Ex. 1011)
`2.
`Brynne investigates the effect of CYP2D6 heterogeneity on the
`pharmacokinetics of tolterodine, as well as potential differences in selected
`pharmacodynamic properties (heart rate, visual accommodation, and
`salivation) of tolterodine as compared to 5-HMT. See Ex. 1011, Abstract.
`Brynne’s study involved “[s]ixteen male subjects (eight extensive
`metabolizers and eight poor metabolizers) [who] received 4 mg tolterodine
`by mouth twice a day for 8 days followed by a single intravenous infusion of
`1.8 mg tolterodine for 30 minutes after a washout period.” Id.
`With respect to the muscarinic side effect dry mouth, Brynne reports
`that “[a] distinct drug effect was [] obtained for four of eight extensive
`metabolizers and most of the poor metabolizers after oral administration. For
`extensive metabolizers, the effect was equally pronounced after intravenous
`compared with oral administration, whereas salivation was less affected
`among poor metabolizers after the infusion.” Id. at 535. In considering the
`relation between the severity of dry mouth and unbound serum levels of the
`two compounds, Brynne reports that “[t]here was a weak correlation between
`tolterodine concentration and effect on salivation. A stronger correlation was
`seen with [5-HMT] and effect.” Id. at 536. Nevertheless, “[o]nly minor
`differences in pharmacodynamic effects after tolterodine dosage were
`observed between the groups. Tolterodine caused a similar decrease in
`salivation in both panels. The decrease occurred when the concentration of
`unbound tolterodine and 5-hydroxymethyl metabolite among extensive
`metabolizers was comparable with that of tolterodine among poor
`metabolizers.” Id., Abstract. Brynne suggests that “the similarity in salivary
`effects between the two phenotypic groups” may be explained by the 10-fold
`
`
`
`12
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`greater serum protein binding of tolterodine as compared to 5-HMT. Id. at
`535–536.
`Brynne also notes a shift in the effect curve with respect to visual
`accommodation with five of the poor metabolizers reporting abnormal visual
`accommodation. Id. at 536, 538. The authors posit that “the most likely
`explanation is the physicochemical differences between tolterodine and
`[5-HMT]. Tolterodine is tenfold more lipophilic than [5-HMT], and
`consequently tolterodine penetrates membranes more rapidly.” Id. at 538.
`Brynne concludes that:
`Despite the influence of CYP2D6 polymorphism on the
`pharmacokinetics of tolterodine, this does not appear to be of
`great pharmacodynamic importance. This is because either high
`concentrations of the parent compound are mainly responsible for
`the effect among poor metabolizers or substantial concentrations
`of the active metabolite [5-HMT] are responsible for the effect
`among extensive metabolizers.
`Id.; see id. at Abstract.
`3.
`Detrol Label (Ex. 1009)
`Detrol Label discusses the structural formula, pharmacokinetics, and
`pharmacology of tolterodine, provided as tolterodine tartrate “for the
`treatment of patients with an overactive bladder with symptoms of urinary
`frequency, urgency, or urge incontinence.” Ex. 1009, 5.13 The reference
`states that:
`Tolterodine is extensively metabolized by the liver following oral
`dosing. The primary metabolic route involves the oxidation of
`the 5-methyl group and is mediated by the cytochrome P450 2D6
`and leads to the formation of a pharmacologically active
`5-hydroxymethyl metabolite [i.e., 5-HMT]. Further metabolism
`
`13 Because the Detrol Label does not include any page numbers, we refer to
`the page numbers Petitioner added to the document.
`
`
`
`13
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`
`leads to formation of the 5-carboxylic acid and N-dealkylated
`5-carboxylic acid metabolites, which account for 51% ± 14% and
`29% ± 6.3% of the metabolites recovered in the urine,
`respectively.
`Id. at 2. Detrol Label notes that about 7% of the population lack cytochrome
`P450 2D6 activity and are designated “poor metabolizers” as compared to the
`general population (“extensive metabolizers”). Id. Pharmacologic studies
`reveal that tolterodine is metabolized at a slower rate in poor metabolizers
`resulting in “significantly higher serum concentrations of tolterodine and
`negligible concentrations of [5-HMT].” Id. But “[b]ecause of differences in
`the protein-binding characteristics of tolterodine and [5-HMT], the sum of
`unbound serum concentrations of tolterodine and [5-HMT] is similar in [both
`populations].” Id. Moreover, “[s]ince tolterodine and [5-HMT] have similar
`antimuscarinic effects, the net activity of DETROL Tablets is expected to be
`similar in extensive and poor metabolizers.” Id.
`In addressing potential drug-drug interactions related to 2D6
`heterogeneity, Detrol Label states that “[t]olterodine is not expected to
`influence the pharmacokinetics of drugs that are metabolized by cytochrome
`P450 2D6 . . . .” Id. at 3. The reference further discloses that fluoxetine is a
`potent inhibitor of cytochrome P450 2D6 activity and has been shown to
`significantly inhibit the metabolism of tolterodine to 5-HMT such that the
`pharmacokinetics of the drug in extensive metabolizers resembles that of
`poor metabolizers. Id. The reference, nevertheless, states that “[n]o dose
`adjustment is required when DETROL and fluoxetine are coadministered.”
`Id. Although Detrol Label does not suggest altering tolterodine dosages for
`2D6 poor metabolizers, because a substantial portion of the drug is
`
`
`
`14
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`N-dealkylated by cytochrome P450 3A4, it recommends dose reduction for
`patients taking drugs that inhibit 3A4. Id. at 2, 5, 7.
`4.
`Bundgaard (Ex. 1012)
`Bundgaard describes prodrug design for drug delivery. Ex. 1012, v.
`According to Bundgaard, “[a] prodrug is a pharmacologically inactive
`derivative of a parent drug molecule that requires spontaneous or enzymatic
`transformation within the body in order to release the active drug, and that
`has improved delivery properties over the parent drug molecule.” Id.
`Bundgaard explains that prodrugs bridge the gap between drug action and
`efficient delivery to a desired target site:
`A molecule with optimal
`structural configuration and
`physicochemical properties for eliciting the desired therapeutic
`response at its target site does not necessarily possess the best
`molecular form and properties for its delivery to its point of
`ultimate action. Usually, only a minor fraction of doses
`administered reaches the target area and, since most agents
`interact with non-target sites as well, an inefficient delivery may
`result in undesirable side effects. This fact of differences in
`transport and in situ effect characteristics for many drug
`molecules is the basic reason why bioreversible chemical
`derivatization of drugs, i.e., prodrug formation, is a means by
`which a substantial improvement in the overall efficacy of drugs
`can often be achieved.
`
`Id.
`
`Bundgaard teaches that esters are popularly used in the design of
`prodrugs because the body contains numerous, widely distributed esterases
`that can cleave such prodrugs to their active forms. Id. at 3–4. With respect
`to parent drugs containing a hydroxyl moiety, exemplary prodrugs have
`employed, for example, carboxylate, carbonate, phosphate, diacetyl, amino
`
`
`
`15
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`acid, ditolulyl, dipivaloyl, aromatic, and hemisuccinate esters. See Id. at 3,
`Table 2.
`Bundgaard further teaches that “[e]ster formation has long been
`recognized as an effective means of increasing the aqueous solubility of
`drugs containing a hydroxyl group, with the aim of developing prodrug
`preparations suitable for parenteral administration.” Id. at 7. This approach
`makes it “feasible to obtain derivatives with almost any desirable
`hydrophilicity or lipophilicity as well as in vivo lability . . . .” Id. at 4. “The
`most commonly used esters for increasing aqueous solubility of alcoholic
`drugs are hemisuccinates, phosphates, dialkylaminoacetates and amino acid
`esters.” Id. at 8.
`5.
`Bundgaard PCT (Ex. 1020)
`Bundgaard PCT describes ester and diester prodrug derivatives of
`morphine for transdermal delivery. Ex. 1020, 1–5, 7–8, 10, 15. In contrast to
`morphine, “the morphine esters [were] more lipophilic than the parent drug in
`terms of octanol-aqueous buffer partition coefficients” and “the 3-hexanoyl,
`3,6-dihexanoyl and other 3,6-dipropionyl morphine esters readily penetrated
`human skin.” Id. at 9–10.
`6.
`Berge (Ex. 1013)
`In a review of pharmaceutical formulation salts, Berge states that:
`The chemical, biological, physical, and economic characteristics
`of medicinal agents can be manipulated and, hence, often
`optimized by conversion to a salt form. Choosing the appropriate
`salt, however, can be a very difficult task, since each salt imparts
`unique properties to the parent compound.
`Salt-forming agents are often chosen empirically. Of the many
`salts synthesized, the preferred form is selected by pharmaceutical
`chemists primarily on a practical basis: cost of raw materials, ease
`of crystallization, and percent yield. Other basic considerations
`
`
`
`16
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`
`include stability, hygroscopicity, and flowability of the resulting
`bulk drug. Unfortunately, there is no reliable way of predicting
`the influence of a particular salt species on the behavior of the
`parent compound. Furthermore, even after many salts of the same
`basic agent have been prepared, no efficient screening techniques
`exist to facilitate selection of the salt most likely to exhibit the
`desired pharmacokinetic, solubility and formulation profiles.
`Ex. 1013, 1. Berge Table I is a list of FDA-approved, commercially
`marketed salts, along with an indication of how frequently those salts were
`used in the pharmaceutics industry as of 1974. Id. at 2. Table I indicates that
`fumarate salts were used 0.25% of the time. Id.
`7.
`Johansson (Ex. 1005)
`Johansson discloses compounds of the general formula I reproduced
`below:
`
`
`Ex. 1005, 1:18–2:4. General formula I represents a class of
`3,3-diphenylpropylamines. Id. at Abstract. In formula I, “R1 signifies
`hydrogen or methyl, R2 and R3 independently signify hydrogen, methyl,
`methoxy, hydroxy, carbamoyl, sulphamoyl or halogen, and X represents a
`tertiary amino group . . . .” Id. at 1:27–30. Johansson further discloses that
`
`
`
`17
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`preferred tertiary amino groups of formula I include the group reproduced
`below:
`
`
`Id. at 2:26–3:5. Johansson teaches that such compounds “can form salts with
`physiologically acceptable acids . . . . Examples of such acid addition salts
`include the hydrochloride, hydrobromide, hydrogen fumarate, and the like.”
`Id. at 2:5–10. According to Dr. Patterson, Johansson’s general formula
`encompasses 5-HMT (Ex. 1003 ¶¶ 133–136), which Patent Owner does not
`contest (see PO Resp. 58 (referencing “Johansson’s disclosure of potential
`salts of 5-HMT”)).
`
`D. Analysis of Alleged Obviousness over Postlind, Bundgaard Publications,
`Detrol Label, and Berge (Ground 1)
`Petitioner asserts that claims 1, 3, 4, and 6–8 would have been obvious
`over the combination of Postlind, Bundgaard, Bundgaard PCT, Detrol Label,
`and Berge. See Pet. 3, 22–38. Petitioner argues that it would have been
`obvious for one of ordinary skill in the art to (1) identify 5-HMT as a lead
`compound for drug development; (2) recognize that 5-HMT could have
`adverse effects due to its metabolism and poor oral bioavailability due to its
`lipophilicity profile; (3) address such concerns regarding adverse effects and
`poor oral bioavailability by esterifying 5-HMT to create a prodrug having
`increased lipophilicity and, subsequently, optimizing the ester moiety to
`arrive at a compound having a short-chain mono-ester derivative at only the
`5-hydroxyl position; and (4) select an acid-addition salt that provides the
`desired product stability. Id. at 22–38. Petitioner also argues that it would
`
`
`
`18
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`have been obvious for one of ordinary skill in the art to treat a patient
`suffering from urinary incontinence with fesoterodine fumarate as
`encompassed by in claims 4, 6, and 7. Id. at 38–40.
`A determination of whether a new chemical compound would have
`been obvious over the prior art typically follows a two prong inquiry
`considering first, whether one of ordinary skill would have selected one or
`more lead compounds for further development and, second, whether the prior
`art would have supplied sufficient motivation to modify a lead compound to
`arrive at the compound claimed with a reasonable expectation of success.
`See Otsuka Pharm. Co., Ltd., v. Sandoz, Inc., 678 F.3d 1280, 1291–92 (Fed.
`Cir. 2012).
`1.
`Identification of 5-HMT as a Lead Compound
`In the first step of our analysis, we determine “whether a chemist of
`ordinary skill would have selected the asserted prior art compounds as lead
`compounds, or starting points, for further development efforts.” Id. at 1291.
`A lead compound comprises “a natural choice for further development
`efforts,” Altana Pharma AG v. Teva Pharm. USA, Inc., 566 F.3d 999, 1008
`(Fed. Cir. 2009), i.e., a prior art compound “that would be most promising to
`modify in order to improve upon its . . . activity and obtain a compound with
`better activity,” Takeda Chem. Indus., Ltd. v. Alphapharm Pty., Ltd., 492
`F.3d 1350, 1357 (Fed. Cir. 2007). “In determining whether a chemist would
`have selected a prior art compound as a lead, the analysis is guided by
`evidence of the compound’s pertinent properties.” Otsuka Pharm., 678 F.3d
`at 1292; see also Eisai Co. v. Dr. Reddy’s Labs., Ltd., 533 F.3d 1353, 1359
`(Fed. Cir. 2008) (stating that even “post-KSR, a prima facie case of
`
`
`
`19
`
`

`

`IPR2016-00517
`Patent 7,985,772 B2
`
`obviousness for a chemical compound still, in general, begins with the
`reasoned identification of a lead compound”).
`Petitioner begins with the proposition that, in light of Postlind and the
`pharmacodynamic information in the Detrol Label, one of ordinary skill in
`the art would recognize that tolterodine was metabolized to an active
`metabolite, 5-HMT, having beneficial properties as compared to the parent
`compound. Pet. 22–24; see Ex. 1003 ¶¶ 40–43, 95–102. Petitioner argues
`that because the references disclose that tolterodine is metabolized to 5-HMT
`by cytochrome CYP2D6, one of ordinary skill in the art would have elected
`to begin with the 5-HMT metabolite in order to avoid the potential for 2D6
`drug-drug interactions or the propensity of 2D6 poor metabolizers to develop
`adverse side effects when using drugs subject to this pathway. Pet. 23–25.
`In particular, Petitioner relies on Postlind, which provides the general caution
`that “[c]linical studies have demonstrated that individuals with reduced
`CYP2D6-mediated metabolism represent a high-risk group in the population
`with a propensity to develop adverse drug effects” and states that a “number
`of drugs” have been identified as “being affected by CYP2D6
`polymorphism.” Ex. 1010, 292. In light of that experience with other drugs
`metabolized via the 2D6 pathway, Postlind suggests that for tolterodine,
`“[t]he possibility of clinical drug interaction at the enzyme level [] exists,
`especially if tolterodine is administered at the same time as a compound that
`is preferentially metabolized by CYP2D6 or to individuals associated with
`the poor CYP2D6 poor metaboli

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