throbber

`
`
`
`
`
`
`
`
`UNITED STATES PATENT AND TRADEMARK OFFICE
`
`
`BEFORE THE PATENT TRIAL AND APPEAL BOARD
`
`
`
`MYLAN PHARMACEUTICALS INC.
`Petitioner,
`
`v.
`
`BRISTOL-MYERS SQUIBB COMPANY and PFIZER INC. ,
`Patent Owners.
`
`
`U.S. Patent No. 9,326,945 to Patel et al.
`
`Inter Partes Review IPR2018-00892
`
`
`
`DECLARATION OF KINAM PARK, PH.D.
`
`
`MYLAN EXHIBIT 1002 
`
`1
`
`

`

`
`
`
`
`
`1.
`
`I, Kinam Park, Ph.D., have been retained by counsel for Petitioner
`
`Mylan Pharmaceuticals Inc. (“Mylan”). I understand that Mylan is petitioning for
`
`inter partes review (“IPR”) of U.S. Patent No. 9,326,945 to Patel et al. (the ’945
`
`patent”) (Ex. 1001), which is assigned to Bristol Myers Squibb Company and
`
`Pfizer Inc. (“Patent Owners”), to request that the United States Patent and
`
`Trademark Office cancel certain claims of the ’945 patent as unpatentable. I
`
`submit this expert declaration in support of Mylan’s IPR petition for the ’945
`
`patent.
`
`I. QUALIFICATIONS AND BACKGROUND
`
`A. Education and Experience
`
`2.
`
`I am the Showalter Distinguished Professor of Biomedical
`
`Engineering and Professor of Pharmaceutics at Purdue University. I am also
`
`President of Akina, Inc., a research company located in West Lafayette that I
`
`founded in 2001, and that focuses on polymers for drug delivery and biomedical
`
`applications as well as developing new drug delivery technologies.
`
`3. My research, training, and experience are all in the areas of
`
`pharmaceutical sciences, specifically in the areas of pharmaceutics,
`
`biopharmaceutics, and biomedical engineering. My research has focused on drug
`
`delivery, including delivery of poorly soluble drugs through use of particle size
`
`reduction to nano/micro drug crystals, and oral formulations of fast-dissolving
`2
`
`

`

`
`
`
`tablets and gastric retention devices, localized drug delivery using drug-device
`
`combination products, and injectable long-acting depot formulations using
`
`biodegradable polymers. My extensive research in these areas includes in vitro
`
`experimentation and in situ and in vivo studies in animals, including evaluating and
`
`analyzing the dissolution and absorption of immediate-release and extended-
`
`release solid oral dosage forms. I have also conducted theoretical analyses and
`
`simulations of drug behavior in the body.
`
`4.
`
`I received a B.S. degree in Pharmacy from Seoul National University
`
`in Seoul, Korea in 1975. I received a Ph.D. degree in Pharmaceutics from the
`
`University of Wisconsin, Madison in 1983. In 1985, I completed my postdoctoral
`
`training in Chemical Engineering at the University of Wisconsin, Madison.
`
`5.
`
`I began working at Purdue University in February 1986. From
`
`February 1986 through June 1994, I was an Associate Professor in the Department
`
`of Pharmaceutics. In July 1994, I became a Professor in the Department of
`
`Pharmaceutics and became a Professor in the Department of Biomedical
`
`Engineering in July 1998. In 2006, I was named the Showalter Distinguished
`
`Professor of Biomedical Engineering at the Weldon School of Biomedical
`
`Engineering, Purdue University, while maintaining a Professor position at the
`
`College of Pharmacy.
`
`6.
`
`As noted above, in 2001, I founded Akina, Inc., which provides a
`
`3
`
`

`

`
`
`
`variety of research products and services with a focus on controlled release and
`
`biomedical applications. Akina, Inc. includes two divisions: PolySciTech, its
`
`products division, and Akinalytics, its laboratory services division. The
`
`PolySciTech division focuses on providing research reagents and products,
`
`including biodegradable/biocompatible polymers, fluorescent products, chemical
`
`intermediates, assay kits, and other products used in biomedical research and
`
`development. Leveraging the developed experience with synthesis and
`
`characterization of novel polymer formulation, as well as in-house research and
`
`development, the Akinalytics division provides an array of services that range from
`
`simple analysis of a single sample to multi-year contracted research.
`
`7.
`
`I have authored or co-authored over 560 publications, comprising
`
`books, book chapters, cover stories, and referred articles in the area of
`
`pharmaceutics, including reducing the particle size of compounds in order to
`
`improve solubility and bioavailability. I have given almost 300 invited lectures. I
`
`hold 22 patents.
`
`8.
`
`I have served on the editorial board of numerous journals in the area
`
`of drug delivery, including, for example, Advanced Drug Delivery Reviews, Drug
`
`Delivery Technology, Encyclopedia of Pharmaceutical Technology, Journal of
`
`Drug Delivery Science and Technology, Nano Reviews, and Drug Delivery and
`
`Translational Research, to name a few. I served as associate editor and book
`
`4
`
`

`

`
`
`
`review editor for Pharmaceutical Research, guest editor for Colloids and Surfaces
`
`B: Biointerfaces, and guest editor for Advanced Drug Delivery Reviews.
`
`Currently, I have been serving as the Editor-in-Chief of the Journal of Controlled
`
`Release since 2005.
`
`9.
`
`I am a member of numerous professional societies, including the
`
`American Association of Pharmaceutical Scientists, American Chemical Society,
`
`Controlled Release Society, the Society for Biomaterials, and Biomedical
`
`Engineering Society.
`
`10.
`
`I have also served on scientific advisory boards for numerous
`
`symposiums, including the International Symposium on Recent Advances in Drug
`
`Delivery System, the Advisory Panel on Excipients: Substance and
`
`Characterization Expert Committee for the United States Pharmacopeia (or USP),
`
`the European Symposium on Controlled Drug Delivery, and the International
`
`Symposium on Microencapsulation.
`
`11. A copy of my curriculum vitae, which lists my publications, patents,
`
`invited lectures, and professional activities and describes my qualifications in
`
`detail, is attached as Exhibit A.
`
`B.
`
`12.
`
`Bases for Opinions
`
`In forming my opinions set forth in this declaration, I have considered
`
`and relied upon my education, background, and decades of experience in the field
`
`5
`
`

`

`
`
`
`of pharmaceutical sciences, including drug delivery, particle size determination
`
`and reduction, dissolution, and formulation science. I have also relied on the
`
`materials listed in Appendix B of the accompanying Petition for Inter Partes
`
`Review by the Petitioner Mylan Pharmaceuticals Inc.
`
`C. Retention and Compensation
`
`13.
`
`I am being compensated for my consulting work on this case at my
`
`rate of $500.00 per hour plus expenses. My compensation in this proceeding is
`
`not dependent on its outcome.
`
`II. LEGAL STANDARDS
`
`14. Counsel has informed me that certain legal principles should guide me
`
`in my analysis. Counsel has informed me that Mylan carries the burden of proving
`
`unpatentability by a preponderance of the evidence, which means Mylan must
`
`show that unpatentability is more likely than not.
`
`15. Counsel has informed me that the question of whether the claims of a
`
`patent are anticipated by, or obvious in view of, the prior art is to be considered
`
`from the perspective of the person of ordinary skill in the art (“POSA”). Counsel
`
`has further informed me that the answer to this question is ascertained as of the
`
`time the invention was made.
`
`16. Counsel has informed me that performing an obviousness analysis
`
`involves ascertaining, as of the time the invention was made, the scope and content
`
`6
`
`

`

`
`
`
`of the prior art, the level of skill of the POSA, the differences between the claimed
`
`invention and the scope and content of the prior art, and whether there are
`
`additional factors present that may argue against a conclusion of obviousness (i.e.,
`
`“secondary considerations”), such as unexpected results attributable to the
`
`invention.
`
`17. Counsel has informed me that an invention may be found obvious:
`
`When there is a design need or market pressure to solve a problem and there are a
`
`finite number of identified, predictable solutions, a POSA has good reason to
`
`pursue the known options within his or her technical grasp. If this leads to the
`
`anticipated success, it is likely the product is not of innovation but of ordinary skill
`
`and common sense. In that instance, the fact that a combination was obvious to try
`
`might show that it was obvious under § 103.
`
`18. Counsel has informed me that a prior art reference anticipates a
`
`claimed invention if the prior art reference disclosed each of the claimed elements
`
`of the invention. A prior art reference not expressly disclosing a claim element
`
`may still anticipate or render obvious the claimed invention, if the missing element
`
`is necessarily present, or inherent, in the reference. The missing element, or
`
`characteristic, is inherent in the reference, if the characteristic is a natural result
`
`flowing from the reference’s explicit disclosure.
`
`19. Counsel has informed me that if a patent claims a composition in
`
`7
`
`

`

`
`
`
`terms of a function, property, or characteristic, and the composition itself is in the
`
`prior art, then the claim may be anticipated or obvious in view of the prior art
`
`reference disclosing the composition.
`
`III. DEFINITION OF A PERSON OF ORDINARY SKILL IN THE ART
`
`20.
`
`I understand that a patent claim is evaluated from the perspective of
`
`a POSA, which I understand is a hypothetical person considered to have the skill
`
`level and knowledge of a particular field related to an alleged invention claimed in
`
`a patent. I further understand that this hypothetical skilled artisan is presumed to
`
`have before him or her all of the relevant prior art. I understand that this
`
`“hypothetical person” can be more than one person or a team of people of different
`
`disciplines. The discussions in this declaration are intended to convey the state of
`
`the art and the knowledge of a POSA generally prior to the earliest priority date of
`
`the patent application that issued as the respective ’945 patent.
`
`21.
`
`In view of the subject matter of the ’945 patent, a POSA as of the
`
`patent’s filing date would typically be a Ph.D.-level (degree or experience)
`
`pharmaceutical scientist with two or more years of experience or a person with a
`
`Master’s degree with five or more years of experience who would be well versed
`
`in the design and release of pharmaceutical dosage forms, and would have a
`
`working understanding of the factors relevant to achieving appropriate
`
`bioavailability of an active pharmaceutical ingredient, particularly a substance
`
`8
`
`

`

`
`
`
`known to have solubility issues.
`
`22.
`
`As of the priority date of the ’945 patent, I have been a POSA as
`
`defined above.
`
`IV.
`
`SUMMARY OF OPINIONS
`
`23. First, a POSA would have understood that the occurrence of a
`
`thromboembolic event is a severe medical concern and can be sudden and
`
`catastrophic. A POSA would have understood that the current anticoagulation
`
`therapies of warfarin and heparin were limited and associated with negative side
`
`effects. Therefore, a POSA would have been motivated to develop new,
`
`alternative anticoagulation drug formulations to warfarin and heparin formulations
`
`and would have understood that such drug(s) would be intended to address or
`
`prevent a sudden thromboembolic disorder and would need to be fast-acting and
`
`bioavailable.
`
`24.
`
`Second, a POSA would have been aware that at the time of filing of
`
`the ’945 patent, the two most promising anticoagulation targets were Factor Xa and
`
`thrombin. However, because of its important role in the coagulation cascade, and
`
`more favorable characteristics over thrombin, a POSA would have been motivated
`
`to target inhibitors of Factor Xa.
`
`25. Third, at the time of filing of the ’945 patent, apixaban was known to
`
`be a potent, specific inhibitor of Factor Xa and was actively being pursued for the
`
`9
`
`

`

`
`
`
`treatment of thromboembolic events in an oral dosage form. A POSA would have
`
`further known that apixaban was polymorphic and that microparticulate, crystalline
`
`forms of apixaban were known and desirable. Therefore, a POSA would have had
`
`more than a reasonable expectation of success from the prior art of making a
`
`crystalline apixaban tablet or capsule formulation containing microcrystalline
`
`apixaban particles. A POSA would have been aware of the research and clinical
`
`development of apixaban and would have been aware that 2.5 mg or 5 mg b.i.d. or
`
`q.d. were identified as efficacious dosages for the treatment and prophylaxis of
`
`thromboembolic events or could have arrived at these dosages using routine
`
`methods of experimentation and optimization.
`
`26. Fourth, although a POSA would have been motivated to develop a
`
`crystalline apixaban oral dosage form as a potent drug candidate for the treatment
`
`and prophylaxis of thromboembolic events, a POSA would have been aware from
`
`Wei and other prior art that apixaban was sparingly soluble, and would have thus
`
`been motivated to pursue avenues to address apixaban’s solubility and dissolution
`
`issues in order to develop the immediate release profile necessary to treat the
`
`serious and often sudden and potentially fatal thromboembolic events.
`
`27. Fifth, a POSA would have understood that reducing particle size is
`
`one of the first avenues to pursue in addressing solubility and dissolution issues of
`
`poorly soluble compounds and would have been aware that a crystalline apixaban
`
`10
`
`

`

`
`
`
`with a reduced particle size addressing these concerns was already disclosed and
`
`claimed by Wei. It would have been a matter of routine optimization for a POSA
`
`to arrive at the claimed particle sizes of the ’945 patent which would result in an
`
`optimum immediate-release dissolution and bioavailability profile. Further, a
`
`POSA would have had a reasonable expectation of success of arriving at the
`
`claimed particle size values because Wei discloses examples where crystalline
`
`apixaban particles were made within the claimed ranges.
`
`28.
`
`Sixth, it is a matter of routine experimentation for a POSA concerned
`
`with improving solubility and dissolution to conduct in vitro tests to determine the
`
`dissolution profile of the crystalline apixaban with a reduced particle size. A
`
`POSA, seeking to develop a drug for eventual FDA approval, would naturally
`
`consult with FDA guidances and would utilize the known recommended methods
`
`for such dissolution tests. A POSA would have recognized that the dissolution
`
`profile is an inherent result or characteristic of the formulation being tested
`
`according to the routine dissolution testing method.
`
`29.
`
`Seventh, all elements recited in the ’945 patent’s dependent claims
`
`were already disclosed in the prior art.
`
`V. BACKGROUND ON THROMBOEMBOLIC EVENTS AND ANTI-
`COAGULANT AGENTS
`30. A thrombosis refers to the formation of one or more blood clots (i.e.,
`
`a thrombus or thrombi) in a blood vessel, and embolism is the blocking of a blood
`11
`
`

`

`
`
`
`vessel by an embolus or emboli that are fragments broken away from a thrombus.
`
`Thus, thromboembolism refers to partial or complete blocking of a blood vessel
`
`caused by embolus or emboli. (See, e.g., “The Surgeon General’s Call to Action to
`
`Prevent Deep Vein Thrombosis and Pulmonary Embolism, U.S. Department of
`
`Health and Human Services, 2008, 7 (Ex. 1018).) Deep vein thrombosis (“DVT”)
`
`is the formation of thrombi in one of the body’s large veins. (Id.) Partial or
`
`complete blocking of an artery or one of its branches in the lungs by thrombi
`
`traveling in the blood stream results in a pulmonary embolism. (Id.) The
`
`occurrence of a thromboembolic event is a major medical concern in that it can
`
`cause serious life-threatening complications. (Id., 9.) The estimated overall annual
`
`incidence of venous thromboembolism (“VTE”) in the United States as of 2010 is
`
`between 1 and 2 per 1000 of the population or 300,000-600,000 cases. (See, e.g.,
`
`Beckman, M.G. et al., “Venous thromboembolism: a public health concern,” Am.
`
`J. Prev. Med., 38(4s):S495-S501, S495 (2010) (Ex. 1019).) The significance of
`
`thromboembolism is not limited to the United States. According to a multinational
`
`report of European Union countries, more than 370,000 VTE-related deaths are
`
`reported per annum, with 34% of these deaths resulting from sudden fatal PE. (See
`
`Cohen, “Venous thromboembolism (VTE) in Europe. The number of VTE events
`
`and associated morbidity and mortality,” Thromb. Haemost., 98(4):756-764 (2007)
`
`(Ex. 1020).) Thus, a POSA understood as of the earliest priority date of the ’945
`
`12
`
`

`

`
`
`
`patent that treatment and prevention of VTE events was clinically important and
`
`would have been motivated to seek fast-acting treatment options.
`
`31. A primary treatment strategy for these disorders is to treat the area or
`
`system already impacted by a blood clot and prevent the clots from reoccurring.
`
`Treatment of VTE events may involve the use of anticoagulants and/or
`
`thrombolytic therapy options, with the use of anticoagulants being the most
`
`common. (See Remko, “Molecular structure, lipophilicity, solubility, absorption,
`
`and polar surface area of novel anticoagulant agents,” J. Mol. Structure, 916:76-85
`
`(2009) (Ex. 1021).) Heparin and warfarin had been the main forms of treatment
`
`for patients with venous and arterial thromboembolic disease for half a century
`
`before the priority date of the ’945 patent. (Ex. 1004, 1937.) Administration of
`
`heparin is hampered because it has to be administered by injection. The
`
`administration of warfarin is also limited, due to a “narrow therapeutic index, slow
`
`onset of therapeutic effect, numerous dietary and drug interactions, and a need for
`
`monitoring as well as dose adjustments.” (Id.) Despite the availability of heparin
`
`or warfarin, thrombotic diseases remain the leading cause of death in developed
`
`countries. Thus, a POSA was further aware of the great clinical need for
`
`alternative oral anticoagulants as a replacement for warfarin and heparin for both
`
`the long-term prevention and treatment of patients with venous and arterial
`
`thromboembolism. (Ex. 1004, 1937-1938, 1944.)
`
`13
`
`

`

`
`
`
`32.
`
`Because of its central location in the coagulation cascade and the fact
`
`that they block both intrinsic and extrinsic pathways, the art recognized that two
`
`favorable targets for the advancement of treatment of thromboembolic disorders
`
`were inhibitors of Factor Xa and thrombin. (Id., 1937.) However, because serine
`
`proteases were recognized to play an important role in coagulation and the
`
`thrombotic process, a POSA was motivated to target Factor Xa over thrombin for
`
`further development. (Id.) This was based on the understanding in the art that
`
`(1) a smaller dose of a Factor Xa anticoagulant is needed to block the coagulation
`
`process earlier than for thrombin; (2) directly inhibiting thrombin may be
`
`associated with a rebound hypercoaguable state not seen with Factor Xa inhibitors;
`
`(3) Factor Xa inhibitors incompletely block thrombin generation and some existing
`
`thrombin in the system has anti-inflammatory functions that may add to an
`
`antithrombotic potential; and (4) in vitro assays suggest that Factor Xa inhibitors
`
`may have a wider therapeutic window than thrombin inhibitors. (Id., 1937-1938.)
`
`33.
`
`Thus, apixaban would have been known by a POSA as an effective
`
`oral anticoagulant. The coagulation cascade pathway is shown below in Figure 1,
`
`and lists many of the agents of the day, including apixaban, and their targeted
`
`coagulation factors:
`
`14
`
`

`

`
`
`
`
`
`(Id., 1938.)
`
`VI. BACKGROUND ON DRUG DELIVERY OF POORLY SOLUBLE
`ORAL DOSAGE FORMS
`34.
`
`In designing a dosage form for administration to a patient, a POSA
`
`generally considers oral dosage forms more desirable over other delivery
`
`mechanisms such as parenteral or intravenous. Oral dosage forms are
`
`advantageous in that they can be easily self-administered by the patient, are
`
`relatively low-cost to manufacture, package and ship, generally have increased
`
`stability, and are virtually tamper resistant. (Ex. 1010, 333.) This is reflective in
`
`that well over 80% of drugs marketed in the United States are oral dosage forms,
`
`with tablets being the most common oral dosage form. (Id.)
`
`35.
`
`The most important role of a drug delivery system is to get the drug
`
`“delivered” to the site of action in a sufficient amount and at the appropriate rate.
`
`15
`
`

`

`
`
`
`(Ex. 1013, 2701.) A POSA further recognizes several factors as potentially
`
`presenting challenges in oral delivery of new drug substances, including drug
`
`absorption and sufficient and reproducible bioavailability and/or pharmacokinetic
`
`profile in humans. (Ex. 1011, Abstract.) Drug absorption from an oral solid
`
`dosage form depends on the release of the drug substance from the drug product,
`
`the dissolution kinetics or solubilization kinetics of the drug under physiological
`
`conditions, and the permeability across the gastrointestinal tract. (Ex. 1015, 1.)
`
`Drug dissolution is a prerequisite for oral absorption, because if the drug is not
`
`fully dissolved it cannot be completely absorbed through the GI epithelium. It is
`
`thus extremely important to understand drug dissolution and solubility in aqueous
`
`media, both in early drug discovery studies and as a prerequisite for the subsequent
`
`formulation development. (See Ungell et al., “Biopharmaceutical Support in
`
`Candidate Drug Selection,” in Pharmaceutical Preformulation and Formulation,
`
`M. Gibson, ed., Interpharm/CRC, 97-153, 100 (2004) (Ex. 1022).) Drug
`
`dissolution/solubility data gives important information that can answer important
`
`questions including regarding whether the drug dissolution/solubility limits the
`
`bioavailability to the extent that it can endanger the usefulness of the drug or
`
`whether the drug substance form should be changed to improve dissolution (e.g.,
`
`changing the salt, polymorph, or particle size). (Id.) The solubility and dissolution
`
`data of a drug further guides whether dissolution rate-enhancing principles (e.g.,
`
`16
`
`

`

`
`
`
`wetting agents, micronisation, solubilizing agents, solid solutions, emulsions, or
`
`nanoparticles) should be applied during the formulation development. (Id.)
`
`36.
`
`Solubility or dissolution in different solvents is an intrinsic
`
`characteristic of the molecule. (Ex. 1011, 250.) Solubility is commonly described
`
`in the unit of mg/mL or a similar unit indicating the concentration of a dissolved
`
`drug. Solubility describes an equilibrium value, and it does not indicate how fast a
`
`drug can be dissolved in an aqueous solution. Dissolution refers to the dynamic
`
`process by which a drug is dissolved in a solvent and is characterized by a rate,
`
`generally the amount dissolved per unit time.
`
`37.
`
`The time to reach equilibrium varies depending on the drug
`
`substance properties, amount of material used, and the equilibrium method used.
`
`(See Tong, W-Q, “Practical aspects of solubility determination in pharmaceutical
`
`preformulation, in Solvent Systems and Their Selection,” in Pharmaceutics and
`
`Biopharmaceutics, P. Augustijns and M. Brewster (eds.), Springer, New York,
`
`137-149, 138 (2007) (Ex. 1023).) Samples generally reach equilibrium reasonably
`
`quick (e.g., if good agitation is use, often within 24 hours). For poorly soluble
`
`drugs, however, the equilibrium time may be unrealistically long due to poor
`
`dissolution rate. (Id.)
`
`38.
`
`Pharmacological activity of a drug is achieved only when it is soluble
`
`in physiological intestinal fluids to be present in the dissolved state at the site of
`
`17
`
`

`

`
`
`
`absorption. (Ex. 1011, 250.) Thus, a drug needs to be released quickly for
`
`absorption, and a poorly soluble drug is usually formulated for fast dissolution.
`
`Aqueous solubility and dissolution kinetics of a drug in intestinal fluids is critical
`
`to bioavailability issues. Dissolution kinetics of a drug can be altered either
`
`through material engineering of the drug substance or through formulation
`
`approaches. (Id.) In addition to aqueous solubility/dissolution kinetics, the
`
`permeability of a drug substance is also critical for oral bioavailability. (Id.)
`
`39.
`
`Poorly soluble drugs, in particular, practically insoluble drugs, pose
`
`many challenges in formulation development. For clinical applications, an
`
`immediate release drug formulation needs to be made to dissolve the drug as soon
`
`as possible for efficient absorption from the stomach and the upper small intestine
`
`for rapid onset of drug activity. The fast dissolution is also critical for the drugs
`
`that decrease the absorption as they move along the GI tract. It is known that in
`
`general a drug reaches the ileum in approximately 85 min. (See Yu et al.,
`
`“Biopharmaceutics classification system: the scientific basis for biowaiver
`
`extensions,” Pharm Res., 19(7): 921-925, 922 (2002) (Ex. 1024).) Thus, it is
`
`necessary to prepare an oral immediate release formulation for treating
`
`thromboembolic disorders that allows dissolution in the GI tract as soon as
`
`possible. For these reasons, there is always a need to improve the dissolution
`
`kinetics of a poorly soluble drug. In general, some in the art have proposed that
`
`18
`
`

`

`
`
`
`solubility in water of ≥ 10 mg/mL in pH range of 1 to 7 is sufficient to avoid
`
`absorption problems, while others suggest that drugs with a solubility in water of
`
`less than 0.1 mg/mL often exhibit dissolution limitations on absorption. (Ex. 1022,
`
`104-105.) Even if the solubility of a drug substance in the dissolution medium is
`
`acceptable, the dissolution rate should be the characteristic of the drug release from
`
`the dosage form rather than the solubility of the drug in the dissolution medium.
`
`(Brown, C.K., “Dissolution method development: an industry perspective,” in
`
`Pharmaceutical Dissolution Testing, J. Dressman and J. Krämer (eds.), Taylor &
`
`Francis, New York, 351-372, 354 (2005) (Ex. 1025).) The bioavailability of a
`
`drug is essentially determined by the dynamics and kinetics of solubility (i.e.,
`
`dissolution rate) of the drug. (See Gong et al., “Principles of solubility,” in Solvent
`
`Systems and Their Selection in Pharmaceuticals and Biopharmaceuticals, P.
`
`Augustijins and M. Brewster (eds.), Springer, New York, 1-27, 23 (2007) (Ex
`
`1026).)
`
`40.
`
`The art recognized that the gold standard for classifying a drug’s
`
`solubility is to refer to the applicable pharmacopeia, such as the United States
`
`Pharmacopoeia (“USP”).
`
`A.
`
`41.
`
`Solubility Classification By the United States Pharmacopoeia
`
`The USP, since at least the 1980s, characterizes solubility into seven
`
`(7) different forms based on its parts of solvent required for one part of solute as
`
`19
`
`

`

`
`
`
`set forth in the Table 1 below:
`
`
`
`(Ex. 1011, 250-251.) As indicated in Table 1, a sparingly soluble drug is defined
`
`as a drug with a solubility range of 10-33 mg/mL, while a very slightly soluble
`
`drug is a drug with a solubility range of 0.1-1 mg/mL, and a practically soluble
`
`drug is a drug with a solubility range less than 0.1 mg/ml. (Id., see also United
`
`States Pharmacopeia XX, 20th Revision, Description and Solubility, United States
`
`Pharmacopeial Convention, Inc., Rockville, MD, 1121 (1980) (Ex. 1027); United
`
`States Pharmacopeia, 27th ed., General Notices and Requirements, United States
`
`Pharmacopeial Convention, Inc., Rockville, MD, 1-12, 9 (2004) (Ex. 1016). As
`
`discussed further herein, the prior art discloses that apixaban has poor solubility.
`
`Such poor solubility is affirmed by the solubility measurement provided by the
`
`applicants in the ’945 patent itself of 40 µg/mL (or 0.04 mg/mL). (Ex. 1001,
`
`1:46.) As can be seen in Table 1, apixaban is classified as practically insoluble,
`
`even when the patent applicants’ own solubility measurement is used.
`
`20
`
`

`

`
`
`
`B.
`
`Enhancing Solubility of Poorly Soluble Drugs Was Known in the
`Art
`
`42. While solubility is intrinsic to the material such that it can only be
`
`influenced by a chemical modification to the drug substance, such as salt complex
`
`formation and prodrug synthesis, dissolution is an extrinsic property that can be
`
`influenced by various chemical and physical means, including complexation,
`
`particle size adjustment, surface properties, solid state modification or
`
`solubilization-enhancing formulation strategies. (Ex. 1011, 251.)
`
`43. When a drug substance is identified as having solubility or
`
`dissolution issues, either in vitro or in vivo, several different formulation strategies
`
`are routinely employed to arrive at the desired drug disposition, as indicated in
`
`Table 5 below:
`
`(Id., 255.) As shown above, one of the first strategies a POSA would take to
`
`investigate improving solubility of a poorly soluble drug is particle size reduction.
`21
`
`
`
`

`

`
`
`
`44.
`
`It has been well known that the dissolution rate, rather than the
`
`saturation solubility, is most often the primary determinant in the absorption
`
`process of a poorly soluble drug. (See, e.g., Ex 1022, 102.) Indeed, since the late
`
`nineteenth century, the dissolution of drugs has been mathematically described by
`
`Noyes-Whitney equation:
`
`(cid:1856)(cid:1829)(cid:1856)(cid:1872)(cid:3404)(cid:1830)(cid:1827)(cid:4666)(cid:1829)(cid:3046)(cid:3398)(cid:1829)(cid:4667)
`(cid:1860)
`
`
`
` where dC/dt is the rate of dissolution of the drug particles, D is the diffusion
`
`coefficient of the drug in solution in the gastrointestinal (GI) fluids, A is the
`
`effective surface area of the drug particles in contact with the GI fluids, h is the
`
`thickness of the diffusion layer around each drug particle, CS is the saturation
`
`solubility of the drug in the solution in the diffusion layer, and C is the
`
`concentration of drug in the GI fluids. (Ex. 1009, 286; Ex. 1022, 101.) Here, it is
`
`noted that the effective surface area of the drug particles (A) indicates the surface
`
`area available for dissolution. (See Diebold, S.M., “Physiological parameters
`
`relevant to dissolution testing: hydrodynamic considerations,” in Pharmaceutical
`
`Dissolution Testing, J. Dressman and J. Kramer (eds.), Taylor & Francis, New
`
`York, 127-191, 129 (2005) (Ex. 1028).)
`
`45. As shown in the Noyes-Whitney equation, the rate of drug
`
`dissolution is directly proportional to the surface area (A) of the drug regardless of
`
`the drug solubility. Increasing the surface area is particularly important for drugs
`22
`
`

`

`
`
`
`that possess poor solubility. (Ex. 1009, 288) Accordingly, the smaller the particle
`
`size, the greater the effective surface area exhibited by a given mass of drug and
`
`the higher the dissolution rate. (Id.; see also Ex. 1013, 2703.) Thus, it has been
`
`well-known for over 100 years that particle size reduction can have a profound
`
`effect on dissolution and is likely to result in increased bioavailability, provided
`
`that the absorption of the drug is dissolution rate limited. (Id.; see also Ex. 1010,
`
`335 (“[f]or many drugs, particularly those for which absorption is limited by the
`
`rate of dissolution, attainment of therapeutic levels may depend on achieving a
`
`small particle size; Ex. 1012, 1:17-19 (“[c]ontrol of crystal size and shape enables
`
`the optimization of the dissolution rate and this may maximize the benefit while
`
`minimizing the side effects.”).) The effect of particle size on the dissolution rate of
`
`sparingly soluble drugs has been demonstrated in many instances at least since
`
`1963 by the superior dissolution rates observed after size reduction. (See, e.g., Ex.
`
`1026, 21-22.)
`
`46.
`
`In addition to particle size, it was well known in the art that
`
`dissolution, and hence absorption, of poorly soluble oral dosage forms could also
`
`be improved through use of excipients, such as surfactants. One of the most
`
`commonly used surfactant is sodium lauryl sulfate, also referred in the art as
`
`sodium dodecyl sulfate, which was recognized in the art as improving the rate of
`
`dissolution of poorly soluble drugs. (See Ex. 1013, 2707; Ex. 1012, 3:32-34; Ex.
`
`23
`
`

`

`
`
`
`1010, 359; Ex. 1022, 141, Tbl. 4.9.) Surfactants, such as sodium lauryl sulfate, can
`
`be used as either as a wetting agent or to solubilize the drug substance at above the
`
`critical micelle concentration (“CMC”). (Ex. 1025, 358.)
`
`C. The Biopharmaceutical Classification System (“BCS”)
`
`47. As stated in sections II(A) and (D) below, both the specification of
`
`the ’945 patent refers to, and the Patent Owners relied upon, the classification of
`
`ap

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