throbber
Trials@uspto.gov
`571.272.7822
`
`
`
`
`
` Paper No. 12
`
` Entered: July 20, 2016
`
`
`
`
`
`
`
`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 RAMA G. ELLURU, KRISTINA M. KALAN, and
`ROBERT A. POLLOCK, Administrative Patent Judges.
`
`POLLOCK, Administrative Patent Judge.
`
`
`
`DECISION
`Institution of Inter Partes Review
`37 C.F.R. § 42.108
`
`
`
`
`
`
`
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`
`
`
`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 have jurisdiction under 35 U.S.C. § 314, which provides that an
`
`inter partes review may not be instituted “unless . . . there is a reasonable
`
`likelihood that the petitioner would prevail with respect to at least 1 of the
`
`claims challenged in the petition.” Upon considering the Petition and the
`
`Preliminary Response, we determine that Petitioner has shown a reasonable
`
`likelihood that it would prevail in showing the unpatentability of claims 1, 3,
`
`4, and 6–8. Accordingly, we institute an inter partes review of those claims.
`
`A.
`
`Related Proceedings
`
`Patent Owner asserts that
`
`UCB and Pfizer Inc. (“Pfizer”), the exclusive licensee 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 filer).
`
`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.
`
`
`
`1 Patent Owner provides, as Exhibit 2001, the District Court’s
`Memorandum finding that the defendants in that proceeding “failed to
`
`2
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`v. Dr. Reddy’s Laboratories, Ltd., No. 1:15-cv-01067 (GMS) (D. Del.).
`
`Paper 8, 2; Prelim. Resp. 7–8.
`
`In addition to the case before us, Petitioner requested institution of
`
`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).
`
`Petitioner also requested institution of 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.
`
`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.” Ex. 1001, Abstract.
`
`
`
`The Specification discloses that “normal urinary bladder contractions
`
`are mediated mainly through cholinergic muscarinic receptor stimulation.”
`
`
`
`present a prima facie case that the asserted claims of the patents-in-suit are
`invalid as obvious.” Ex. 2001, 19; see Prelim. Resp. 7–8. Although the
`district court reached this determination on a different record and applying
`different standards, the arguments and references applied overlap with those
`before us. See Ex. 2001, Prelim. Resp. 1–2, 15–17, 21, 25, 33. Accordingly,
`while we are not bound to these findings, we find the court’s analysis
`informative.
`
`3
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`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.
`
`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 55–59 (citing Nilvebrant et al.,
`
`1997, Eur. J. Pharmacol. 327 (1997), 195–207). “WO 94/1 1337 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:
`
`
`
`
`
`4
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`See, e.g., Pet. 19; Ex. 1010, 289; Ex. 1011, 530. As illustrated above,
`
`tolterodine has a single hydroxyl group at the 2-position of the methylated
`
`phenolic ring, whereas 5-HMT bears a second hydroxyl moiety on the
`
`5-position methyl group of that ring.
`
`C.
`
`Challenged Claims
`
`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;
`
`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; Prelim. Resp. 1–2, 7; Ex. 1024, 8, 19.
`
`5
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`D.
`
`The Asserted Grounds of Unpatentability
`
`Petitioner challenges claims 1, 3, 4, and 6–8 of the ’772 patent on two
`
`grounds (Pet. 3, 32–40):
`
`References
`
`Postlind,2 “Bundgaard
`publications,”3,4,5 Detrol
`Label,6 and Berge7
`Brynne,8 Bundgaard, and
`Johansson9
`
`
`Basis
`
`§ 103
`
`Claims Challenged
`
`1, 3, 4, and 6–8
`
`§ 103
`
`1, 3, 4, and 6–8
`
`Petitioner relies also on the Declaration of its technical expert Steven
`
`E. Patterson, Ph.D. (Ex. 1003) and the Declaration of DeForest McDuff,
`
`
`
`2 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”).
`3 We interpret Petitioner’s reference to “Bundgaard publications” as
`referring to Exhibits 1012 and 1020. See Pet. iv, 3, 28,
`4 Bundgaard, Design of Prodrugs Elsevier (1985). Ex. 1012
`(“Bundgaard”).
`5 WO 92/08459, published May 29, 1992. Ex. 1020 (“Bundgaard
`PCT”).
`6 Detrol™ (tolterodine tartrate tablets) prescribing information (1998).
`Ex. 1009 (“Detrol Label”).
`7 Berge et al., Pharmaceutical Salts, 66(1) J. Pharm. Sci. 1–19 (1977).
`Ex. 1013 (“Berge”).
`8 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”).
`9 Johansson et al., WO 94/11337, published May 26, 1994. Ex. 1005
`(“Johansson”).
`
`6
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`Ph.D. (Ex. 1033) with respect to lack of commercial success.10 Patent
`
`Owner relies on the Declaration of William R. Roush, Ph.D. (Ex. 2002).
`
`A.
`
`Claim Construction
`
`
`
`ANALYSIS
`
`In an inter partes review, claim terms in an unexpired patent are
`
`interpreted according to their broadest reasonable constructions in light of
`
`the Specification of the patent in which they appear. See 37 C.F.R.
`
`§ 42.100(b); Cuozzo Speed Techs., LLC v. Lee, No. 15–446, 2016
`
`WL 3369425, at *12 (U.S. June 20, 2016) (upholding the use of the broadest
`
`reasonable interpretation standard). Under the broadest reasonable
`
`construction standard, claim terms are presumed to have their ordinary and
`
`customary meaning, as would be understood by one of ordinary skill in the
`
`art in the context of the entire disclosure. In re Translogic Tech., Inc., 504
`
`F.3d 1249, 1257 (Fed. Cir. 2007).
`
`In the present case, neither party contests the meaning of any claim
`
`term. See Pet. 6; Prelim. Resp. 9. Only those terms that are in controversy
`
`need be construed, and 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). For purposes of this decision, we determine that no
`
`claim term requires express construction.
`
`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
`
`
`
`10 Patent Owner does not assert commercial success in the Preliminary
`Response.
`
`7
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`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
`
`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.
`
`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. The level of ordinary skill in the art is further demonstrated
`
`8
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`by the prior art asserted in the Petition. See Okajima v. Bourdeau, 261 F.3d
`
`1350, 1355 (Fed. Cir. 2001).
`
`C. Overview of the Asserted References
`
`We begin our discussion with a brief summary of the references
`
`asserted.
`
`i. Postlind (Ex. 1010)
`
`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)
`
`[i.e., 5-HMT]” by cytochrome P450 2D6 (“CYP2D6” or 2D6”) “and
`
`dealkylation of the nitrogen” by cytochrome P450 CYP3A4 (“CYP3A4”).
`
`
`
`9
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`Id. at 289; see also id. at 292 (concluding that the dealkylation reaction “is
`
`predominantly catalyzed by CYP3A4 in human liver microsomes.”)11
`
`
`
`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
`
`poor 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.
`
`ii. Brynne (Ex. 1011)
`
`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
`
`
`
`11 Petitioner’s technical expert, Dr. Patterson, emphasizes that 5-HMT
`is also N-dealkylated by CYP3A4. See Ex. 1003 ¶¶ 111, 45–46 (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. 287–295 (1997). See Ex. 1007
`(“Brynne 1997”), 291 Fig. 2.
`
`10
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`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 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. The authors posit that “the most likely explanation is the
`
`11
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`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 pharmacodynamics 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., Abstract.
`
`iii. 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 overactive bladder with symptoms of urinary
`
`frequency, urgency, or urge incontinence.” Ex. 1009, 5. The reference
`
`states that:
`
`Tolterodine is extensively metabolized in 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
`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
`
`12
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`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, a
`
`potent inhibitor of cytochrome P450 2D6 activity, 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 dosage
`
`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
`
`N-dealkylated by cytochrome P450 3A4, it recommends dose reduction for
`
`patients taking drugs that inhibit 3A4. Id. at 2, 5, 7.
`
`iv. Bundgaard (Ex. 1012)
`
`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.” Ex. 1012,
`
`13
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`v. 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 when 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 drugs containing a hydroxyl moiety, exemplary prodrugs have employed,
`
`for example, carboxylate, carbonate, phosphate, diacetyl, amino acid,
`
`ditoluluyl, 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 hydrophobicity as well as in vivo lability.” Id. at 4. “The
`
`most commonly used esters for increasing aqueous solubility of alcoholic
`
`14
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`drugs are hemisuccinates, phosphates, dialkylaminoacetates and amino acids
`
`esters.” Id. at 8.
`
`v. Bundgaard PCT (Ex. 1020)12
`
`Bundgaard PCT describes ester and diester prodrug derivatives of
`
`morphine for transdermal delivery. Ex. 1020, 2–5, 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.
`
`vi. 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 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.
`
`
`
`12 Petitioner variously refers to Exhibit 1020 as “Bundgaard PCT,”
`“Bundgaard Patent,” and, collectively with Ex. 1012, “Bundgaard
`publications.” See Pet. iv, 3, 28,
`
`15
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`Ex. 1013, 1. Berge Table I is a list of FDA-approved, commercially
`
`marketed salts, along with an indication of how frequently those salts are
`
`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.
`
`vii. 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, hydroxyl, carbamoyl, sulphamoyl or halogen, and X represents a
`
`tertiary amino group.” Id. at 1:27–30. Johansson further discloses that
`
`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 salt
`
`forms 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.
`
`16
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`D. Obviousness Analysis
`
`Petitioner challenges claims 1, 3, 4, and 6–8 of the ’772 patent on two
`
`grounds. Pet. 3. Patent Owner argues that Petitioner has not presented
`
`sufficient data or objective evidence to show a reasonable likelihood that it
`
`would prevail in showing the unpatentability of claims 1, 3, 4, and 6–8.
`
`Prelim. Resp. 11–41. We present an overview of the parties’ positions
`
`below.
`
`i. Ground I: Obviousness over Postlind, Bundgaard Publications,
`Detrol Label, and Berge
`
`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. To briefly summarize
`
`Petitioner’s argument, 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 would have poor oral
`
`bioavailability due to its lipophilicity profile; (3) address these concerns
`
`regarding 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.
`
`1. Identification of 5-HMT
`
`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, 99, 101. Petitioner argues
`
`17
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`that because these references disclose that tolterodine is metabolized to
`
`5-HMT by cytochrome P450 2D6, 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 this 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 metabolizer phenotype.” Id.
`
`Patent Owner responds that Postlind’s caution regarding the
`
`possibility of clinical drug interactions related to 2D6 metabolism is
`
`superseded by, e.g., the teachings of the Detrol Label, Brynne, and
`
`Nilvebrandt 1997,13 which collectively teach that tolterodine and 5-HMT
`
`have “almost identical” pharmacological profiles; that “[d]espite the
`
`influence of CYP2D6 polymorphism on the pharmacokinetics of tolterodine,
`
`
`
`13 Nilvebrant et al., Antimuscarinic Potency and Bladder Selectivity of
`PNU-200577, a Major Metabolite of Tolterodine, 81 PHARMACOLOGY &
`TOXICOLOGY 169–172 (1997). Ex. 1015.
`
`18
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`this does not appear to be of great pharmacodynamics importance”; that
`
`“[t]olterodine is not expected to influence the pharmacokinetics of drugs that
`
`are metabolized by cytochrome p450 2D6”; and that no dose adjustment is
`
`required when tolterodine is co-administered with the potent 2D6 inhibitor
`
`fluoxetine. See Prelim. Resp. 12–16; Ex. 1015, 172; Ex. 1011, 538;
`
`Ex. 1009, 2. Thus, Patent Owner argues, “[i]n view of the prior art as a
`
`whole, a person of skill in the art would have had no reason to avoid
`
`tolterodine’s CYP2D6 polymorphism and would have had no reason to turn
`
`to 5-HMT.” Prelim. Resp. 12–13 (citation omitted).
`
`Although we find Patent Owner’s arguments reasonable, Patent
`
`Owner’s “supporting evidence concerning disputed material facts will be
`
`viewed in the light most favorable to the petitioner for purposes of deciding
`
`whether to institute an inter partes review.” 37 C.F.R. § 42.108(b).
`
`Accordingly, on the present record, we find that Petitioner has demonstrated
`
`a reasonable likelihood that one of ordinary skill in the art would have
`
`selected 5-HMT over tolterodine for further development.
`
`2. Reason to Modify 5-HMT
`
`With respect to a reason to modify 5-HMT, Petitioner first states that
`
`Postlind “would have motivated a person of ordinary skill to modify 5-HMT
`
`to a compound that avoided CYP2D6 metabolism as known to occur with
`
`tolterodine.” Pet. 23. Insofar as 5-HMT is the result of CYP2D6
`
`metabolism of tolterodine (i.e., there is no evidence that 5-HMT undergoes
`
`CYP2D6 metabolism), we do not find this statement persuasive.
`
`Petitioner also focuses on the Detrol Label’s dose reduction
`
`recommendation for “patients with significantly reduced hepatic function or
`
`who are currently taking drugs that are inhibitors of cytochrome P450 3A4”
`
`but, as noted in section II(C)(i), tolterodine and 5-HMT are both P450 3A4
`
`19
`
`

`

`Case IPR2016-00517
`Patent 7,985,772 B2
`
`substrates. See Pet. 24–25. Petitioner has not explained adequately how
`
`Detrol Label’s concerns relating the P450 3A4 substrate profile of
`
`tolterodine would have motivated one of ordinary skill in the art to modify
`
`either compound.
`
`Petitioner also relies on paragraphs 116 and 118 of Dr. Patterson’s
`
`report in asserting that “a person of ordinary skill in the art would have
`
`appreciated that 5-HMT was too lipophilic and needed to be modified in a
`
`way to improve bioavailability.” Pet. 27. We presume that Petitioner’s
`
`assertion that 5-HMT was “too lipophilic” is intended as an argument that 5-
`
`HMT is not lipophilic enough. See id. at 10 (asserting that 5-HMT was
`
`known to have “poor lipophilicity”). Relevant to Ground I, Dr. Patterson
`
`asserts in paragraph 112 of his report that
`
`[w]hen the skilled artisan would have looked at 5-HMT, it
`would have seen that the presence of two hydroxyl groups around
`th

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