throbber
UNITED STATES PATENT AND TRADEMARK OFFICE
`
`BEFORE THE PATENT TRIAL AND APPEAL BOARD
`
`MYLAN PHARMACEUTICALS INC.
`
`Petitioner
`
`V.
`
`ASTRAZENECA AB
`
`Patent Owner
`
`Case IPR2016—01326
`
`U.S. Patent No. 8,466,139
`
`DECLARATION OF RONALD J. SAWCHUK, Ph.D. IN SUPPORT OF
`PATENT OWNER’S PRELIMINARY RESPONSE
`
`AstraZeneca Ex. 2003 p. 1
`Mylan Pharms. Inc. V. AstraZeneca AB IPR2016-01326
`
`

`

`I.
`
`INTRODUCTION
`
`1.
`
`I have been retained by AstraZeneca AB (“Astraleneca”) in
`
`connection with this mterpartes review proceeding (IPR20l6-01326) before the
`
`United States Patent and Trademark Office Patent Trial and Appeal Board
`
`(“Board”).
`
`2.
`
`I understand that Mylan Pharmaceuticals Inc. (“Mylan”) has
`
`challenged AstraZeneca-owned U.S. Patent No. 8,466,139, which relates to a
`
`method of treating hormonal dependent disease of the breast or reproductive tract,
`
`and, more specifically, hormonal dependent breast cancer.
`
`3.
`
`I further understand that Mylan has petitioned institution of this inter
`
`partes review proceeding on the basis of several references identified in its Petition
`
`(IPR20l6-01326, Paper 1) (“Petition”).
`
`4.
`
`I am being compensated $750 per hour for my time consulting in this
`
`matter.
`
`I have no financial interest in the outcome of this proceeding and my
`
`compensation is in no way contingent upon my opinions or the outcome of this
`
`proceeding.
`
`II.
`
`QUALIFICATIONS
`
`5 .
`
`I am a Professor of Pharmaceutics, Emeritus, and Morse Alumni
`
`Distinguished Teaching Professor at the University of Minnesota.
`
`I also served as
`
`the Director of the Bioanalytic and Pharmacokinetic Services Laboratory at the
`
`AstraZeneca Ex. 2003 p. 2
`
`

`

`University of Minnesota until August of 2014.
`
`I have studied and carried out
`
`clinical and pre-clinical research in the field of pharmacokinetics and
`
`biopharmaceutics for over forty years.
`
`6.
`
`I joined the University of Minnesota in 1971 as an Instructor in
`
`Pharmaceutics after having obtained a Bachelor and Masters of Science Degree
`
`from the University of Toronto in 1963 and 1996, respectively, and completing my
`
`Doctoral Degree (Ph.D.) in Pharmaceutical Chemistry (pharmacokinetics
`
`emphasis) at the University of California, San Francisco, which was granted in
`
`1972.
`
`7.
`
`At the University of Minnesotal served as an Assistant Professor of
`
`Pharmaceutics from 1972 to 1977, an Associate Professor of Pharmaceutics from
`
`1977 to 1983, and a full Professor of Pharmaceutics from 1983 until my retirement
`
`in July of 2010. During this period, I was course director for instruction in
`
`pharmacokinetics, clinical pharmacokinetics, advanced pharmacokinetics, and
`
`pharmacokinetic modeling and simulation.
`
`1 was also a participating instructor in
`
`biopharmaceutics, and advanced pharmacokinetics, 1 continue to provide lectures
`
`relating to preclinical and clinical pharmacokinetics to scientists in the
`
`pharmaceutical industry.
`
`8.
`
`I also served as a member of the graduate programs in Pharmaceutics,
`
`Neurosciences, and Experimental and Clinical Pharmacology. From 1983 to 1989
`
`AstraZeneca Ex. 2003 p. 3
`
`

`

`and 1991 to 1994, I was the Director of Graduate Studies in Pharmaceutics at the
`
`University. From 1982 to 1995, I also served as Director of the Clinical
`
`Pharmacokinetics Laboratory at the College of Pharmacy at the University of
`
`Minnesota. From 1998 to 1999 I served as the Head of the Department of
`
`Pharmaceutics at the University of Minnesota.
`
`9.
`
`Although I have formally retired from the University, my Graduate
`
`Faculty appointment in the Department of Pharmaceutics is still in effect, allowing
`
`me to teach graduate students in the program.
`
`I have advised on the order of forty
`
`graduate students, postdoctoral fellows, and visiting scholars, on projects relating
`
`to preclinical and clinical pharmacokinetics, biopharrnaceutics, and bioanalytical
`
`chemistry.
`
`10. A major focus of my research was preclinical and clinical
`
`pharmacokinetics. I have been involved with many different preclinical and
`
`clinical human trials, and in particular with the analysis of the pharrnacokinetic and
`
`other data generated during those trials.
`
`1 also focused my research on drug
`
`bioavailability and bioequivalence. I have taught, and continue to teach,
`
`pharmacokinetics, and pharmacokinetic modeling and simulation in professional,
`
`graduate, and elective courses at the University of Minnesota and to the
`
`pharmaceutical industry. This instruction includes lectures on the assessment of
`
`bioavailability and bioequivalence.
`
`AstraZeneca Ex. 2003 p. 4
`
`

`

`11.
`
`I have expertise in the determination of pharmacokinetic parameters
`
`and metrics for orally administered drugs, bioanalytical chemistry,
`
`biopharmaceutics, and pharmacodynamics. I have devoted a large part of my
`
`career to the study of the pharmacokinetics of drugs. And, in addition to authoring
`
`numerous publications in this area, I have received funding from various sources in
`
`the public and private sector to support my research in pharmacokinetics, including
`
`support from the National Institutes of Health (“NIH”) and the U.S. Food and Drug
`
`Administration (“FDA”).
`
`12. During my career, I received several honors, scholarships and awards,
`
`including the Weaver Medal of Honor in 2001, the Meritorious Manuscript Award
`
`from the American Association of Pharmaceutical Scientists in 1999 and the Hallie
`
`Bruce Memorial Lecture Award in 1996. In 2007, I received the American
`
`Pharmacists Association (APhA) Research Achievement Award in the Basic
`
`Pharmaceutical Sciences.
`
`13.
`
`I have been a member of numerous scientific and clinical societies.
`
`I
`
`am a Fellow of the American Association of Pharmaceutical Scientists and of the
`
`American Association for the Advancement of Science.
`
`I have been a member of
`
`the International Society of Anti-infective Pharmacology and the International
`
`Society for the Study of Xenobiotics (IS SX).
`
`I served a three-year term as a
`
`AstraZeneca Ex. 2003 p. 5
`
`

`

`member-at-large on the American Association of Pharmaceutical Scientists
`
`(AAPS) Executive Council.
`
`14.
`
`I have served on the editorial boards of scientific journals such as the
`
`Journal of Pharmaceutical Sciences.
`
`I am currently on the Editorial Board of the
`
`AAPS Journal, and on the IS SX Journal, Xenobiotica.
`
`I have also served on
`
`numerous advisory committees and review panels.
`
`15.
`
`I am a named author on over 100 refereed scientific publications,
`
`several book chapters and over 170 abstracts, which have been presented at
`
`scientific meetings. I have also co-edited a book on drug bioavailability and given
`
`hundreds of invited lectures.
`
`16.
`
`I have significant experience in the areas of pharmaceutical research,
`
`pharmacokinetics, and drug development. Therefore, I believe that I am qualified
`
`to render the opinions set forth in this declaration.
`
`17. My academic background and work experience are summarized in my
`
`curriculum vitae, attached to this declaration as Exhibit A.
`
`III.
`
`ISSUES CONSIDERED
`
`18.
`
`In this declaration, I was asked to provide opinions concerning:
`
`i.
`
`The qualifications of a person of ordinary skill in the art as of
`
`January 10, 2000;
`
`ii.
`
`The state of the art as of January 10, 2000;
`
`AstraZeneca Ex. 2003 p. 6
`
`

`

`iii.
`
`The construction of the claim terms “wherein the method
`
`achieves a blood plasma fulvestrant concentration of at least 2.5
`
`ngm1'1 for at least two weeks” (independent Claims 1 and 11)
`
`and “wherein the blood plasma fulvestrant concentration is at
`
`least 8.5 ngml_1” (dependent Claims 4 and 14) and “wherein the
`
`blood plasma fulvestrant concentration is attained for at least
`
`four [4] weeks” (dependent Claims 6, 10, 16 and 20); and
`
`iv.
`
`The declaration of Dr. Laird Forrest, Ph.D. (Ex. 1003) (“Forrest
`
`Decl.”).
`
`IV. MATERIALS CONSIDERED
`
`19.
`
`In preparing this declaration, I reviewed the IPR Petition submitted by
`
`Mylan; Howell 1996 and McLeskey (Ex. 1005 and Ex. 1006, respectively); the
`
`’139 Patent (Ex. 1001); the declaration of Dr. Forrest (Ex. 1003); and the other
`
`exhibits listed in Exhibit B.
`
`V.
`
`SUMMARY OF APPLICABLE LEGAL CONSIDERATIONS
`
`20.
`
`Counsel for AstraZeneca requested that I express my opinions with
`
`certain guidelines in mind, which are set forth below.
`
`21.
`
`For this declaration I have been asked to use January 10, 2000 as the
`
`relevant date for my analysis.
`
`AstraZeneca Ex. 2003 p. 7
`
`

`

`22. AstraZeneca’s counsel informed me that my analysis must be done
`
`through the eyes of the “person of ordinary skill in the art” as of January 10, 2000.
`
`I understand from AstraZeneca’s counsel that a person of ordinary skill in the art is
`
`a hypothetical person, who has the characteristics of an ordinary artisan including
`
`ordinary creativity.
`
`23.
`
`Factually, in my opinion, a person of ordinary skill in the art in 2000
`
`would have been a person having a bachelor’s or advanced degree in a discipline
`
`such as pharmacy, pharmaceutical sciences, endocrinology, medicine or related
`
`disciplines, and having at least two years of practical experience in drug
`
`development and/or drug delivery, or the clinical treatment of hormonal dependent
`
`diseases of the breast and/or reproductive tract. Because drug formulation and
`
`development is complicated and multidisciplinary, it would require a team of
`
`individuals including, at least, medical doctors, formulators and
`
`pharmacokineticists.
`
`24. Unless I expressly state otherwise, all of the opinions provided in this
`
`declaration are made from the perspective of a person of ordinary skill in the art as
`
`of January 10, 2000.
`
`AstraZeneca Ex. 2003 p. 8
`
`

`

`VI.
`
`THE STATE OF THE ART AS OF JANUARY 10, 2000
`
`A.
`
`Drug Delivery And Pharmacokinetics
`
`25. Drug targeting and duration of delivery are two important aspects of
`
`drug delivery. Drug targeting concerns identifying a specific organ or tissue to
`
`which the drug is to be delivered, while duration of delivery refers to how long the
`
`drug is present in the target organ or tissue.
`
`26. Here, the point of the formulations set forth in the challenged patent
`
`claims is to deliver specified blood plasma levels of the drug fulvestrant for
`
`specified times.
`
`27.
`
`In terms of duration, one conventional distinction involves the
`
`difference between immediate and sustained released formulations.
`
`28.
`
`“Immediate release” means the active pharmaceutical ingredient is
`
`released without a delay from its dosage form after it is administered. Most
`
`conventional oral formulations, such as tablets or capsules, are designed for
`
`immediate release of active pharmaceutical ingredients upon administration in
`
`order to rapidly obtain complete absorption.
`
`29.
`
`Characteristic of immediate release formulations is a relatively rapid
`
`rise in the blood plasma drug levels—to an early and high peal<—followed by a
`
`relatively rapid decrease in those levels.
`
`AstraZeneca Ex. 2003 p. 9
`
`

`

`30.
`
`In contrast, sustained-release formulations are characterized by a
`
`relatively slow rise in blood plasma drug levels which peak later, and are followed
`
`by a relatively prolonged decrease in those levels. These formulations are also
`
`often referred to as extended-release formulations.
`
`31. With “sustained release” “blood level oscillation characteristic of
`
`multiple dosing of conventional dosage forms is reduced, because a more even
`
`blood level is maintained.” Ex. 2134 (Lachman’s) at 5. “Sustained-release
`
`systems include any drug delivery system that achieves slow release of drug over
`
`an extended period of time. . .The objective in designing a sustained-release system
`
`is to deliver drug at a rate necessary to achieve and maintain a constant drug blood
`
`level.” Ex. 2080 (Remington’s) at 6.
`
`32. Without question, a person of ordinary skill would have understood
`
`that a “sustained-release” formulation is “designed to achieve a prolonged
`
`therapeutic effect by continuously releasing medication over an extended period of
`
`time after administration of a single dose. In the case of inj ectable dosage forms,
`
`this period may vary from days to months.” Ex. 2134 (Lachman’s) at 5. In other
`
`words, sustained release formulations are “designed to achieve a prolonged
`
`therapeutic effect by continuously releasing medication over an extended period of
`
`time after administration of a single dose.” Id.
`
`AstraZeneca Ex. 2003 p. 10
`
`

`

`33. Many sustained-release formulations are described in terms of a
`
`specific minimum drug concentration (“at least concentration X”) that is achieved
`
`and maintained over a particular period of time (eg, hours, a day, a week, two
`
`weeks, a month).
`
`34.
`
`The study of the time-course of blood plasma levels of a drug
`
`following administration of a particular formulation/active pharmaceutical
`
`ingredient is called pharmacokinetics. Amongst other things, pharmacokinetics
`
`offers a means by which to compare the rate and extent of drug exposure provided
`
`by different formulations and/or dosing of the same active pharmaceutical
`
`ingredient.
`
`35.
`
`This rate and extent of drug exposure requires in viva pharmacokinetic
`
`studies. In a clinical study setting, pharmacokinetists determine the concentration
`
`of drug in a subject’s plasma over time (by periodically drawing blood) in order to
`
`understand how the body processes the drug as it is being absorbed from a given
`
`formulation after it has been administered. Typically a graph of plasma drug
`
`concentrations as a function of time is generated. This graph is referred to as a
`
`“concentration-time course” or “concentration-time curve.” And, a variety of
`
`analytical methods can then be used to study the results.
`
`36.
`
`The figure below illustrates the difference between the time-course of
`
`a sustained-(solid curve) and immediate-release (dotted curve) formulation for a
`
`AstraZeneca EX. 2003 p. ll
`
`

`

`single dose. In contrast to an immediate-release formulation, the sustained-release
`
`formulation exhibits a prolonged period during which plasma concentrations are
`
`maintained in a specified range (e.g., above some minimum effective level).
`
`immediate-release
`
`
`
`drugconcentrationinplasma
`
`_ _______-.,_____________________ \ minimum effective level
`
`sustained-release
`
`/’
`
`time after dosing
`
`B.
`
`Pharmacokinetics, Pharmacodynamics, And The Development 01'
`Drugs Through Clinical Trials
`
`37.
`
`Clinical trials are conducted in a series of steps, referred to as Phases.
`
`If a drug is found to be “successful” in a given Phase, it is permitted to continue to
`
`the next. Typically there are three such Phases, referred to as Phase I, H and 111,
`
`respectively.
`
`38.
`
`The disciplines of pharmacokinetics and pharmacodynamics are
`
`important areas of activity throughout clinical development.
`
`39.
`
`Pharmacokinetics involves studying the relationship between the dose
`
`and/or dosing regimen used and the resulting plasma or serum concentrations of
`
`the drug. The plasma concentration-time profile or levels observed depend upon
`
`AstraZeneca Ex. 2003 p. 12
`
`

`

`the rate and extent of absorption of the drug from its dosage form into the
`
`bloodstream, in addition to how it is distributed within the body, and how rapidly
`
`and efficiently it is clear from the body by the organs of elimination (e.g., kidney
`
`and/or liver).
`
`40.
`
`Related to pharmacokinetics are bioavailability and bioanalytical
`
`chemistry. Bioavailability is a measure of the rate and extent of absorption of a
`
`drug into systemic blood, in animals or humans. The extent of absorption is
`
`typically characterized by the area under the curve (“AUC”) in the blood plasma
`
`following either a single dose or upon multiple dosing over a specified duration.
`
`The rate of absorption is usually characterized by the maximum concentration of
`
`the drug observed in plasma, and the time at which this maximum is observed.
`
`These parameters or metrics are referred to as “Cmax” and “Tmax,” respectively.
`
`Bioanalytical chemistry involves the quantitative analysis of biological fluids (e.g.,
`
`plasma, whole blood, urine, and cerebrospinal fluid) for endogenous, e.g.,
`
`hormones, and exogenous compounds, e.g., drugs and metabolites. This field
`
`includes the measurement and analysis of drug levels in plasma, which provides
`
`data used to calculate many pharmacokinetic parameters or metrics, such as AUC,
`
`Cmax, and Tmax.
`
`41. Of note, systemic exposure to a drug may be described in terms of the
`
`blood serum or plasma concentrations of the drug during continuous therapy (e.g.,
`
`AstraZeneca Ex. 2003 p. 13
`
`

`

`the steady-state plasma concentration, CSS), or the area under the blood plasma
`
`concentration-time curve (the “area under the curve”, or AUC).
`
`42.
`
`Pharmacodynamics involves the study of the potential relationship
`
`between plasma levels of a drug and the biological effects produced. These
`
`include both the desired therapeutic responses (efficacy) and side effects or adverse
`
`events. Although efficacy (the desired therapeutic response) may be linked to
`
`plasma levels, this relationship is often very difficult to identify for a variety of
`
`reasons including the complex and usually unknown mechanisms of action for
`
`many drugs. Other complicating factors include both the potential disequilibrium
`
`in the concentrations of a drug at the measurement site (i.e. the blood plasma or
`
`serum) with those in what is referred to as the “effect compartment” and the
`
`cascade of events that may need to occur over time before a response to the drug is
`
`observed.
`
`43.
`
`Significant data (usually including data from Phase III clinical trials)
`
`and a careful analysis of the relationship between plasma drug levels and the
`
`effects that a drug produces is required to establish any “pharmacokinetic—
`
`pharmacodynamic” link.
`
`C.
`
`Targeted Blood Plasma Drug Concentrations During Therapy
`
`44.
`
`If a relationship between plasma concentrations and response-
`
`efficacy and/or adverse effects—can be established for a drug, that may allow for
`
`AstraZeneca EX. 2003 p. 14
`
`

`

`the development of a strategy involving achieving and maintaining a target
`
`concentration or a target range of concentrations for individual patients.
`
`45.
`
`This target(s) corresponds with the greatest likelihood of therapeutic
`
`success. Stated differently, ranges of serum or plasma concentrations of a drug
`
`which are known to be therapeutically significant can be used prospectively to
`
`establish a closing regimen for patients.
`
`46.
`
`It may be important to monitor plasma concentrations in individual
`
`patients during therapy if one wishes to ensure that those levels are within the
`
`therapeutic range, in particular if for some reason the patient’s medical condition
`
`or genomic class warrants it. However, this is not always necessary, for example,
`
`if the field’s experience with the drug product and dosing regimen has established
`
`the typical blood plasma drug concentrations obtained.
`
`VII. CLAIM CONSTRUCTION
`
`47.
`
`I have been asked to consider Claims 1, 4, 6, 10, 11, 14, 16 and 20 of
`
`the ’139 Patent, and specifically the limitations directed to blood plasma
`
`fulvestrant concentrations and durations.
`
`Claiml
`
`A method for treating a hormonal dependent benign or malignant
`disease of the breast or reproductive tract comprising administering
`intramuscularly to a human in need of such treatment a formulation
`comprising:
`about 5 0 mgml'1 of fulvestrant,
`a mixture of from 17-23% w/v of ethanol and benzyl alcohol;
`12-18% w/v of benzyl benzoate, and
`
`AstraZeneca Ex. 2003 p. 15
`
`

`

`a sufficient amount of castor oil vehicle;
`wherein the method achieves a blood plasma fulvestrant concentration
`of at least 2.5 ngml’1 for at least two weeks.
`
`Claim 4
`
`Claim 6
`
`The method of claim 1, wherein the blood plasma fulvestrant
`concentration is at least 8.5 ngml_1.
`
`The method of claim 1, wherein the blood plasma fulvestrant
`concentration is attained for at least four weeks.
`
`Claim 10 The method of claim 3, wherein .
`. the blood plasma fulvestrant
`.
`concentration is attained for at least four weeks.
`
`Claim 11 A method for treating a hormonal dependent benign or malignant
`disease of the breast or reproductive tract comprising administering
`intramuscularly to a human in need of such treatment a formulation
`consisting essentially of:
`about 50 mgml'1 of fulvestrant;
`a mixture of from 17-23% w/v of ethanol and benzyl alcohol;
`12-18% w/v of benzyl benzoate; and
`a sufficient amount of castor oil vehicle;
`wherein the method achieves a blood plasma fulvestrant concentration
`of at least 2.5 ngml'1 for at least two weeks.
`
`Claim 14 The method of claim 11, wherein the blood plasma fulvestrant
`concentration is at least 8.5 ngmlil.
`
`Claim 16 The method of claim 11, wherein the blood plasma fulvestrant
`concentration is attained for at least four weeks.
`
`Claim 20 The method of claim 13, wherein .
`. the blood plasma fulvestrant
`.
`concentration is attained for at least four weeks.
`
`48.
`
`In my opinion, the “wherein” clauses of these claims are essential in
`
`order to practice the claims.
`
`49.
`
`The claimed methods do not specifically set forth the dose of
`
`fulvestrant that is to be administered nor the frequency of administration. Rather,
`
`these claims allow for both to be varied so long as the resultant blood plasma
`
`AstraZeneca EX. 2003 p. 16
`
`

`

`fulvestrant concentration and duration limitations are satisfied—that is more than a
`
`desired result, it is the result that must be obtained in order to practice the claims.
`
`50. Moreover, these blood plasma fulvestrant concentration and duration
`
`limitations characterize and distinguish the claimed methods as involving the use
`
`of a “sustained release pharmaceutical formulation.” Ex. 1001 , at abstract.
`
`51.
`
`As explained above, a sustained release formulation slowly releases
`
`drug over an extended period of time to achieve and maintain a prolonged
`
`therapeutic effect (also often described as an extended release formulation). Supra
`
`at W 30-33, 36.
`
`52.
`
`This stands in contrast to conventional or immediate release
`
`formulations, which result in a rapid rise in blood plasma drug levels—to an early,
`
`high peak—followed by a relatively rapid decrease in those levels. Supra at M 28-
`
`29, 36.
`
`53.
`
`The objective of a sustained release formulation is completely
`
`different than that for an immediate release formulation: “[t]he objective in
`
`designing a sustained—release system is to deliver drug at a rate necessary to
`
`achieve and maintain a constant drug blood level” for an extended period of time.
`
`Supra at 1] 31. Moreover, again, as can be seen from the image below, a sustained
`
`release formulation exhibits a completely different concentration-time course than
`
`an immediate release formulation.
`
`AstraZeneca Ex. 2003 p. 17
`
`

`

`immediaterelease
`
`
`
`drugconcentrationinplasma
`
`_ _______*-.,._____________________ \ minimumeffective level
`
`sustained-release
`
`K
`
`time after dosing
`
`54.
`
`It is clear that the invention here involves use of “sustained release”
`
`formulations—a characteristic now firmly set forth in the wherein clauses that
`
`would be removed from the claims if those clauses were to be ignored. As such,
`
`from the perspective of a pharmacokinetist, it makes no sense to ignore those
`
`clauses.
`
`55.
`
`The inventors repeatedly identify “the invention” as concerning a
`
`“sustained release” formulation. For example, in the Abstract the invention is
`
`described as a “novel sustained release pharmaceuticalformulation adapted for
`
`administration by injection containing the compound 7ot-[9-(4,4,5 ,5 ,5 -
`
`pentafluoropentylsulphinyl)nonyl]oestra-l ,3 ,5 (1 O)-triene-3 , l 7 [3-diol, more
`
`particularly to a formulation adapted for administration by injection containing the
`
`compound 70L-[9-(4,4,5 ,5 ,5 -pentafluoropentylsulphinyl)nonyl]oestra- l ,3 ,5 (l 0)-
`
`triene-3,l7B-diol in solution in a ricinoleate vehicle which additionally comprises
`
`at least one alcohol and a non-aqueous ester solvent which is miscible in the
`
`AstraZeneca Ex. 2003 p. 18
`
`

`

`ricinoleate vehicle.” Ex. 1001, at abstract (emphasis added). The “Field of the
`
`Invention” states: “[t]he invention relates to a novel sustained release
`
`pharmaceutical formulation adapted for administration by injection containing the
`
`compound 70L- [9-(4 ,4,5 ,5 ,5 -pentafluoropentylsulphinyl)nonyl]oestra- 1 ,3 ,5 (1 0)-
`
`triene-3,17B-diol.” Ex. 1001, at col.1, 11. 20-23.
`
`5 6.
`
`The sustained release properties and characteristics of the formulation
`
`are, in fact, discussed throughout the specification. For example, the specification
`
`explains that the inventors “surprisingly found that the .
`
`.
`
`. formulations of the
`
`invention provide, after intra-muscular injection, satisfactory release of
`
`fulvestrant over an extended period of time.” Ex. 1001, at col.8, 11.34-36.
`
`(emphasis added). Additionally, the inventors explain “[w]e have found that
`
`despite the rapid elimination of the additional solubilizing excipients .
`
`.
`
`. from the
`
`formulation vehicle and the site of injection after injection of the formulation,
`
`extended release at therapeutically significant levels1 of fulvestrant over an
`
`extended period can still be achieved by the formulation of the invention.” Ex.
`
`1 Of note, the inventors also state, “[b]y use of the term ‘therapeutically significant
`
`levels’ we mean that blood plasma concentrations of at least 2.5 ngml_1, ideally at
`
`least 3 ngml_1, at least 8.5 ngml_1, and up to 12 ngml_1 of fulvestrant are achieved
`
`in the patient.” Ex. 1001, at col.9, ll.1-4.
`
`AstraZeneca Ex. 2003 p. 19
`
`

`

`1001, at col.8, 11.61-67 (emphasis added). Further, the formulation is taught to
`
`achieve a “particularly even release profile,”
`
`1001, at col.10, 11.30-31
`
`(emphasis added)), which as explained above is decidedly not what is achieved
`
`using an immediate release formulation. Supra 1111 28-33, 36.
`
`57.
`
`The patent specification even defines “extended release”: “[b]y use of
`
`the term ‘extended release’ we mean at least two weeks, at least three weeks,
`
`and, preferably at least four weeks of continuous release offulvestrant is
`
`achieved.
`
`In a preferred feature extended release is achieved for 36 days.
`
`Preferably extended release offulvestrant is for at least 2-5 weeks and more
`
`preferablyfor tliefollowingperiods (weeks) 2.5-5, 2.5-4, 3-4, 3.5-4 and most
`
`preferably for at least about 4 weeks.” Ex. 1001, at col.9, 11.6-13 (emphasis
`
`added).
`
`58.
`
`Consistent with the specification, the formulation set forth in the
`
`method of use claims is likewise described as a sustained release formulation
`
`throughout the prosecution history. For example:
`
`i.
`
`Ex. 2132 (Dec. 3, 2002 Office Action), at 6-7 (noting the claim
`
`limitations missing from the prior art, including the limitations
`
`found in the wherein clause: “Dukes does not expressly teach
`
`the plasma concentration offulvestrant herein”) (emphasis
`
`added);
`
`AstraZeneca Ex. 2003 p. 20
`
`

`

`EX. 2133 (Aug. 21, 2008 Amendment), at 8-9 (“The invention
`
`therefore addresses the objective of defining (a) a
`
`pharmaceutically acceptable solvent or mixture of solvents (b)
`
`that will dissolve a sufficient quantity of fulvestrant [at least 250
`
`mg] (c) to form a small enough volume of formulation that is
`
`acceptable for injection [6 ml or less] and will provide ((1) a
`
`fulvestrant concentration of at least 45 mgml'1 [claim 36] and/or
`
`(e) the sustained release of fulvestrant whereby a therapeutically
`
`significant bloodplasmafulvestrant concentration ofat least 2.5
`
`ngml'1 is attainedfor at least 2 weeks [claim 35]”) (emphasis
`
`added);
`
`iii.
`
`Ex. 2133 (Aug. 21, 2008 Amendment), at 14 (“The invention is
`
`focused in particular on the discovery of a novel and unobvious
`
`formulation for this extremely difficult to formulate molecule,
`
`which formulation is suitable for intramuscular injection to a
`
`human patient and is capable of dissolving the therapeutic target
`
`amount of fulvestrant in a small enough volume for IM
`
`administration, and which formulation provides for the
`
`satisfactory sustained release of fulvestrant over an extended
`
`AstraZeneca EX. 2003 p. 21
`
`

`

`period oftime as speczfea’ in the present claims”) (emphasis
`
`added);
`
`iv.
`
`Ex. 2133 (Aug. 21, 2008 Amendment), at 15 (“However,
`
`developing an IM injectable formulation for fulvestrant that
`
`would achieve the satisfactory sustained release of the drug
`
`over an extendedperiod ofweeks presented a particularly
`
`difficult challenge to the experienced formulator”) (emphasis
`
`added);
`
`Ex. 2135 (Aug. 21, 2008 Declaration), at 4 (explaining the goal
`
`of the research was “to formulate an intramuscular (IM)
`
`injection that would provide for the satisfactory sustained
`
`release of fulvestrant over a period of at least two weeks and
`
`preferably over a period of at least four weeks to reduce the
`
`frequency of administration”) (emphasis added);
`
`vi.
`
`Ex. 2141 (Jan. 17, 2012 Amendment), at 5 (“The Office
`
`acknowledges that McLeskey does not expressly teach ‘the use
`
`of fulvestrant in treating hormonal dependent diseases of
`
`breast’, ‘the dosing regimen to be once a month, intramuscular
`
`administration’, ‘the volume administered’, or ‘the herein
`
`claimed serum concentration offu1vestram”’) (emphasis added);
`
`AstraZeneca EX. 2003 p. 22
`
`

`

`vii.
`
`Ex. 2141 (Jan. 17, 2012 Amendment), at 15, 19 (“The POSITA
`
`would not have had a reasonable expectation that the McLes/cey
`
`castor oil composition would have been eflective to administer
`
`fulvestrant intramuscularly to achieve a therapeutic eflectfor at
`
`leastfour weeks, as instantly recited. Two independent reasons
`
`are set forth below supporting a lack of expectation of success
`
`for the combination of the references cited by the Office. .
`
`. [a]
`
`POSITA would not have had an expectation that the results
`
`from subcutaneous injection in McLeskey would have been
`
`applicable to the intramuscular administration of fulvestrant .
`
`.
`
`.
`
`[and n]umerous variables affect the efficacy of an intramuscular
`
`formulation (e.g.,. [sic] identity and proportion of cosolvents)
`
`and a POSITA would have understood that the resulting
`
`variability precludes a POSITA from having an expectation a
`
`priori that a given formulation would be successful in a given
`
`method of treatment until actual suitable in vivo experiments are
`
`performed”) (emphasis added);
`
`viii.
`
`Ex. 213 8 (Jan. 17, 2012 Declaration), at 20-21 (“Moreover, the
`
`examples above underscore the fact that efficacy of a given drug
`
`administered by a given route of dosing (e.g., intramuscular)
`
`AstraZeneca Ex. 2003 p. 23
`
`

`

`cannot be known until appropriate comparative studies are
`
`performed in a suitable animal model. For some drugs, the
`
`desired effect might be achieved following a particular route of
`
`dosing, but for other drugs it might not. The rate and extent of
`
`drug absorption, and the associated pharmacodynamics (e. g., the
`
`achievement of a desired drug effect) may differ greatly
`
`depending on the properties of the drug, the choice of an animal
`
`model, and the site of drug administration. Consequently, one
`
`of ordinary skill in the art having the very limited experimental
`
`subcutaneous data from McLeskey would not have had an
`
`expectation that the intramuscular administration of fulvestrant
`
`using the McLeskey castor oil composition would have been
`
`effective following intramuscular administration, such as in the
`
`method described in the claims. This is especially true because
`
`McLeskey did not disclose plasma or blood levels offulvestrant
`
`in mice after subcutaneous administration of the formulation,
`
`nor any information regarding the rate and/or extent of
`
`absorption offulvestrantfrom the subcutaneous injection site.
`
`Additionally, the claims recite achieving a given therapeutic
`
`plasma concentration for at leastfour weeks, and there is no
`
`AstraZeneca Ex. 2003 p. 24
`
`

`

`information in any of the references cited in the Ojfce Action
`
`that would have suggested that such long-term eflicacy
`
`associated with a single dose would be exhibited using the
`
`McLeskey castor oil composition by any route of administration.
`
`Thus, one of ordinary skill in the art would not have had an
`
`expectation that the castor oil composition disclosed in
`
`McLeskey, which was administered subcutaneously to mice,
`
`would have been therapeutically effective upon intramuscular
`
`administration of fulvestrant, for example, by following the
`
`method described in the claims”) (emphasis added); and
`
`ix.
`
`Ex. 2138 (Jan. 17, 2012 Declaration), at 22-23 (“Regardless of
`
`how high or low the cosolvent concentrations are in a given
`
`formulation, the preparation of formulations in which a drug
`
`such as fulvestrant can be solubilized is not sufficient to ensure
`
`the desired therapeutic effect when such formulation is
`
`administered to patients. As explained in the ’887 application
`
`‘[s]imply solubilising fulvestrant in an oil based liquid
`
`formulation is not predictive of a good release profile or lack of
`
`precipitation of drug after injection at the injection site.’ Exhi

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