throbber
UNITED STATES PATENT AND TRADEMARK OFFICE
`
`
`
`BEFORE THE PATENT TRIAL AND APPEAL BOARD
`
`
`
`INNOPHARMA LICENSING, LLC
`
`Petitioner
`
`V.
`
`ASTRAZENECA AB
`
`Patent Owner
`
`
`
`Case IPR2017-00904
`
`US Patent 6,774,122
`
`
`DECLARATION OF RONALD J. SAWCHUK, PH.D. IN SUPPORT OF
`PATENT OWNER’S PRELIMINARY RESPONSE
`
`AstraZeneca Exhibit 2003 p. 1
`InnoPharma Licensing LLC V. AstraZeneca AB IPR2017-00904
`Fresenius—Kabi USA LLC V. AstraZeneca AB IPR2017-01910
`
`

`

`I)
`
`11)
`
`III)
`
`IV)
`
`V)
`
`VI)
`
`VII)
`
`VIII)
`
`INTRODUCTION........................................................................................ 1
`
`QUALIFICATIONS AND EXPERIENCE ................................................. 1
`
`MY UNDERSTANDING OF THE PROCEEDING ................................... 5
`
`MATERIALS CONSIDERED .................................................................... 6
`
`MY OPINIONS AND THEIR BASES ........................................................ 6
`
`SUMMARY OF APPLICABLE LEGAL CONSIDERATIONS ................ 7
`
`CLAIM CONSTRUCTION ......................................................................... 8
`
`THE STATE OF THE ART AS OF JANUARY 10, 2000 ....................... 10
`
`A)
`
`B)
`
`Drug Delivery And Pharmacokinetics ............................................. 10
`
`Pharmacokinetics, Pharmacodynamics, And The
`Development Of Drugs Through Clinical Trials ............................. 13
`
`C)
`
`Targeted Blood Plasma Drug Concentrations During Therapy ...... 16
`
`IX)
`
`OVERVIEW OF THE PROSECUTION HISTORY ................................ 18
`
`A)
`
`B)
`
`The Sawchuk Declaration Describes Numerous Differences
`That EXist Between McLeskey and the Patent Claims .................... 18
`
`The Gellert Declaration and the Sawchuk Declaration Are
`Consistent and Both Support the Patentability of the
`Challenged Claims ........................................................................... 20
`
`X)
`
`REFERENCES CITED BY DR. BERGSTROM ...................................... 30
`
`A)
`
`Howell (EX. 1007) ........................................................................... 31
`
`B) McLeskey (EX. 1008) ...................................................................... 37
`
`C)
`
`O’Regan (EX. 1009) ......................................................................... 40
`
`XI)
`
`THE CLAIM LIMITATIONS TO BLOOD PLASMA
`
`FULVESTRANT CONCENTRATIONS ARE NOVEL AND NOT
`
`OBVIOUS .................................................................................................. 41
`
`A)
`
`B)
`
`Therapeutically Significant Blood Plasma Fulvestrant
`Concentrations Are Not Taught or Suggested in the Prior Art ....... 41
`
`There Was No Motivation to Combine the Prior Art Cited and
`in Any Event, There Would Have Been No Reasonable
`Expectation of Success in Doing So ................................................ 47
`
`AstraZeneca Exhibit 2003 p. 2
`
`

`

`C)
`
`The Skilled Artisan Would Not Have Expected the Castor
`Oil-Based Formulation in McLeskey to Achieve the Claimed
`Therapeutically Significant Fulvestrant Concentration
`Limitations ....................................................................................... 50
`
`XII)
`
`THE GAPS IN DR. BERGSTROM’S ANALYSIS ARE NOT
`FILLED BY INNOPHARMA’S OTHER EXPERTS’ OPINIONS .......... 54
`
`XIII) CONCLUSION .......................................................................................... 76
`
`AstraZeneca Exhibit 2003 p. 3
`
`

`

`I, Ronald J. Sawchuk, Ph.D., do hereby make the following declaration:
`
`I)
`
`INTRODUCTION
`
`1.
`
`2.
`
`I am over the age of eighteen and competent to make this declaration.
`
`I have been retained as an expert witness on behalf of AstraZeneca
`
`AB for the above-captioned inter partes review (IPR).
`
`I am being compensated at
`
`my customary rate of $875 per hour for my consultation in connection with this
`
`matter. My compensation is in no way dependent on the outcome of my analysis
`
`or opinions rendered in this matter.
`
`II) QUALIFICATIONS AND EXPERIENCE
`
`3.
`
`My name is Ronald J. Sawchuk, PhD.
`
`I am a Professor of
`
`Pharmaceutics, Emeritus, and Morse Alumni Distinguished Teaching Professor.
`
`I
`
`also served as the Director of the Bioanalytic and Pharmacokinetic Services
`
`Laboratory at the University of Minnesota until August of 20 14 when I completed
`
`a drug development contract at the University.
`
`I have studied and carried out
`
`clinical and pre-clinical research in the field of pharmacokinetics and
`
`biopharrnaceutics for over forty years.
`
`4.
`
`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 (PhD) in Pharmaceutical Chemistry (pharmacokinetics
`
`AstraZeneca Exhibit 2003 p. 4
`
`

`

`emphasis) at the University of California, San Francisco, which was granted in
`
`1972.
`
`5.
`
`At the University of Minnesota I 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
`
`biopharrnaceutics, and advanced pharmacokinetics.
`
`I continue to provide lectures
`
`relating to preclinical and clinical pharmacokinetics to scientists in the
`
`pharmaceutical industry.
`
`6.
`
`I also served as a member of the graduate programs in Pharmaceutics,
`
`Neurosciences, and Experimental and Clinical Pharmacology. From 1983 to 1989
`
`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
`
`Pharrnacokinetics 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.
`
`7.
`
`Although I have formally retired from the University, my Graduate
`
`Faculty appointment in the Department of Pharmaceutics is still in effect, allowing
`
`AstraZeneca Exhibit 2003 p. 5
`
`

`

`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, biopharmaceutics, and bioanalytical
`
`chemistry.
`
`8.
`
`A major focus of my research was preclinical and clinical
`
`pharmacokinetics.
`
`l have been involved with many different preclinical and
`
`clinical human trials, and in particular with the analysis of the pharmacokinetic and
`
`other data generated during those trials.
`
`1 also focused my research on drug
`
`bioavailability and bioequivalence.
`
`l 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.
`
`9.
`
`l have expertise in the determination of pharmacokinetic parameters
`
`and metrics for orally administered drugs, bioanalytical chemistry,
`
`biopharrnaceutics, and pharrnacodynamics.
`
`l 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
`
`AstraZeneca Exhibit 2003 p. 6
`
`

`

`Administration (“FDA”).
`
`10. 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.
`
`11.
`
`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 (ISSX).
`
`I served a three-year term as a
`
`member-at-large on the American Association of Pharmaceutical Scientists
`
`(AAPS) Executive Council.
`
`12.
`
`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 ISSX Journal, Xenobiotica.
`
`I have also served on
`
`numerous advisory committees and review panels.
`
`13.
`
`I am a named author on over 100 refereed scientific publications,
`
`several book chapters and over 170 abstracts, which have been presented at
`
`AstraZeneca Exhibit 2003 p. 7
`
`

`

`scientific meetings.
`
`I have also co-edited a book on drug bioavailability and given
`
`hundreds of invited lectures.
`
`14.
`
`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.
`
`15. My academic background and work experience are summarized in my
`
`curriculum vitae, attached to this declaration as Exhibit A.
`
`16.
`
`In the past four years, I have testified in the following litigation:
`
`Ferrmg v. Watson (July 17, 2013); Ferrmg v. Watson and Apotex (Jan 21-30,
`
`2014), Shire v. Actavis et al. (Feb 13, 2014); Astra-Zeneca v. Sandoz et al. (June 5,
`
`2015), EMS v. Teva (August 25, 2015); Astra-Zeneca v. Sandoz et al. (March 23,
`
`2016), and Astra-Zeneca v. Sagent and Glenmark (July 11-14, 2016).
`
`III) MY UNDERSTANDING OF THE PROCEEDING
`
`17.
`
`I have been informed that this proceeding is an inter partes review
`
`(“IPR”) before the Patent Trial and Appeal Board of the United States Patent and
`
`Trademark Office (“the Board”).
`
`I have been informed that an IPR is a proceeding
`
`to review the patentability of one or more issued claims in a United States patent
`
`on the grounds that the patent is the same as or rendered obvious in view of the
`
`prior art.
`
`18.
`
`I understand that InnoPharma Licensing, LLC (“InnoPharma”) has
`
`AstraZeneca Exhibit 2003 p. 8
`
`

`

`challenged AstraZeneca-owned U. S. Patent No. 6,774,122, which relates to a
`
`method of treating hormonal dependent disease of the breast or reproductive tract,
`
`and, more specifically hormonal dependent breast cancer.
`
`19.
`
`l have been informed that InnoPharma filed a Petition (IPR2017-
`
`00904, Paper 1) (“Petition”) requesting IPR of US. Patent No. 6,774,122 (the
`
`122 Patent”), which issued to John R. Evans and Rosalind U. Grundy on August
`
`10, 2004 and is assigned to AstraZeneca AB.
`
`1 have reviewed the Petition, and
`
`understand that it alleges that claims 1, 2, 5 and 9 of the ’ 122 Patent are
`
`unpatentable over Howell 1996 (Ex. 1007) and, alternatively, over the combination
`
`of Howell 1996 (Ex. 1007) with McLeskey (Ex. 1008), and the combination of
`
`Howell 1996 (Ex. 1007) with McLeskey (Ex. 1008) and O’Regan (Ex. 1009).
`
`IV) MATERIALS CONSIDERED
`
`20.
`
`In preparing this declaration, I reviewed the Howell 1996 (Ex. 1007),
`
`McLeskey (Ex. 1008) and O’Regan (EX. 1009), the ’12 Patent (EX. 1001), the
`
`declaration of Dr. Bergstrom (Ex. 1013), and the other exhibits listed in Exhibit B.
`
`V) MY OPINIONS AND THEIR BASES
`
`21.
`
`In this declaration, l was asked to provide opinions concerning:
`
`A.
`
`The qualifications of a person of ordinary skill in the art as of
`
`January 10, 2000;
`
`B.
`
`The state of the art as of January 10, 2000;
`
`AstraZeneca Exhibit 2003 p. 9
`
`

`

`C.
`
`The claim limitations “whereby a therapeutically significant
`
`blood plasma fulvestrant concentration of at least 2.5 ngml‘1 is
`
`attained for at least 2 weeks after injection” (Claims 1 and 2),
`
`and
`
`D.
`
`The declaration of Dr. Richard Bergstrom, PhD. (Ex. 1013)
`
`(“Bergstrom Decl.”) and exhibits cited therein.
`
`22.
`
`As part of this opinion, I considered the level of ordinary skill in the
`
`art around January 2000, which represents the filing date of GB 0000313, to which
`
`the ’ l22 Patent claims priority.
`
`23.
`
`Based on my review of the materials identified above in Section IV)
`
`as well as the materials listed in Exhibit B, and my knowledge and experience, my
`
`opinions are as follows:
`
`VI)
`
`SUMMARY OF APPLICABLE LEGAL CONSIDERATIONS
`
`24.
`
`Counsel for AstraZeneca requested that I express my opinions with
`
`certain guidelines in mind, which are set forth below.
`
`25.
`
`For this declaration I have been asked to use January 10, 2000 as the
`
`relevant date for my analysis.
`
`26. 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
`
`AstraZeneca Exhibit 2003 p. 10
`
`

`

`a hypothetical person, who has the characteristics of an ordinary artisan including
`
`ordinary creativity.
`
`27.
`
`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.
`
`28. Unless expressly stated 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.
`
`VII) CLAIM CONSTRUCTION
`
`29.
`
`All of the claims of the ’ 122 Patent are expressly directed to methods
`
`of treatment. The methods of treatment include choice of an active ingredient, a
`
`method of administration (i.e., a combination of excipients and active injected
`
`intramuscularly), and the amount of the active to be delivered to the blood in a
`
`sustained release fashion to treat hormonal dependent disease of the breast and
`
`AstraZeneca Exhibit 2003 p. 11
`
`

`

`reproductive tract.
`
`30.
`
`I agree with Dr. Bergstrom that the therapeutically significant blood
`
`plasma fulvestrant concentration terms in the ’ 122 Patent claims are limitations of
`
`the claims. Bergstrom Decl. 11 60. These limitations are in claims 1 and 2:
`
`“whereby a therapeutically significant blood plasma fulvestrant concentration of at
`
`least 2.5 ngml‘1 is attained for at least 2 weeks after injection.”
`
`31. A person of skill in the art would understand this limitation to mean
`
`that the specified blood plasma fulvestrant concentrations of at least 2.5 nng'1 is
`
`achieved and maintained for the specified amount of time, i.e., at least 2 weeks.
`
`This is consistent with the Board’s finding in Mylan Pharmaceuticals Inc. v.
`
`AstraZeneca AB, Case IPR2016-01325, Paper No. 11 (P.T.A.B. Dec. 14, 2016) .
`
`EX. 1011 (PTAB Decision) at 18 (“[W]e interpret ‘achieves’ in the wherein clauses
`
`as meaning that the concentration of fulvestrant in a patient’s blood plasma is at or
`
`above the specified minimum concentration for the specified time period”).
`
`32.
`
`Further, these limitations give meaning to and provide defining
`
`characteristics of the method of treatment. Indeed, as the Board previously held,
`
`“rather than merely stating the result of intramuscularly administering the recited
`
`formulation, [] the wherein clause dictates both the administration duration and
`
`dose of the formulation, i.e., an amount sufficient to provide a therapeutically
`
`significant blood plasma fulvestrant concentration of at least 2.5 ngml'1 for at least
`
`AstraZeneca Exhibit 2003 p. 12
`
`

`

`four weeks.” EX. 1011 at 17. And, “[t]hat these parameters are further limited in
`
`claim[] 2, [] (‘the therapeutically significant blood plasma fulvestrant
`
`concentration is at least 8.5 nng'1 ’) further indicates that the wherein clauses
`
`provide defining characteristics.” Id.
`
`VIII) THE STATE OF THE ART AS OF JANUARY 10, 2000
`
`A)
`
`Drug Delivery And Pharmacokinetics
`
`33. 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.
`
`34. 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.
`
`35.
`
`In terms of duration, one conventional distinction involves the
`
`difference between immediate and sustained released formulations.
`
`36.
`
`“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.
`
`10
`
`AstraZeneca Exhibit 2003 p. 13
`
`

`

`37.
`
`Characteristic of immediate release formulations is a relatively rapid
`
`rise in the blood plasma drug levels—to an early and high peak—followed by a
`
`relatively rapid decrease in those levels.
`
`38.
`
`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.
`
`39. 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.
`
`40. 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 injectable 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
`
`ll
`
`AstraZeneca Exhibit 2003 p. 14
`
`

`

`therapeutic effect by continuously releasing medication over an extended period of
`
`time after administration of a single dose.” Id.
`
`41. 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 (e.g., hours, a day, a week, two
`
`weeks, a month).
`
`42.
`
`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.
`
`43.
`
`This rate and extent of drug exposure requires in vivo pharrnacokinetic
`
`studies. In a clinical study setting, pharmacokineticists 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.
`
`12
`
`AstraZeneca Exhibit 2003 p. 15
`
`

`

`44.
`
`The figure below illustrates the difference between the time-course of
`
`a sustained-(solid curve) and immediate-release (dotted curve) formulation for a
`
`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
`
`/
`
`concentrationinplasma
`drug
`
`sustained-release
`
`/
`
`time after dosing
`
`B)
`
`Pharmacokinetics, Pharmacodynamics, And The Development Of
`Drugs Through Clinical Trials
`
`45.
`
`Climcal trials are conducted in a series of steps, referred to as Phases.
`
`If a drug is found to be “successfu ” in a given Phase, it is pemiitted to continue to
`
`the next. Typically there are three such Phases, referred to as Phase I, II and 111,
`
`respectively.
`
`46.
`
`The disciplines of pharmacokinetics and pharrnacodynamics are
`
`important areas of activity throughout clinical development.
`
`47.
`
`Pharmacokinetics is essentially the study of the relationship between
`
`13
`
`AstraZeneca Exhibit 2003 p. 16
`
`

`

`the dose, or dosing regimen that is used in animals or humans, and the plasma or
`
`serum concentrations of the drug that are produced. The profile of plasma
`
`concentrations or levels observed depends upon the rate and extent of absorption of
`
`the drug from its dosage form into the subject’s 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.
`
`48.
`
`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,
`
`CM. and Tmax.
`
`l4
`
`AstraZeneca Exhibit 2003 p. 17
`
`

`

`49. 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.,
`
`the steady-state plasma concentration, Css), or the area under the blood plasma
`
`concentration-time curve (the AUC).
`
`50.
`
`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. For example, a disequilibrium in the concentrations of the drug at the
`
`measurement site (i.e. the blood plasma or serum) and those in what is referred to
`
`as an “effect compartment” often complicates the relationship between drug levels
`
`and therapeutic response. In this case, there may be a significant delay in the
`
`response, such that effects occur much later than expected based upon the pattern
`
`of plasma levels of the drug. Other examples include a situation where there is a
`
`complicated cascade of events that must occur over time (e.g., changes in the level
`
`and activity of clotting factors resulting from the administration of an anticoagulant
`
`that interferes with the “clotting cascade”) before a response to the drug is
`
`observed.
`
`15
`
`AstraZeneca Exhibit 2003 p. 18
`
`

`

`51. A careful analysis of the relationship between plasma drug levels and
`
`the effects that the drug produces is required to establish any “pharmacokinetic-
`
`pharmacodynamic” link and typically requires careful and detailed observations of
`
`the results of numerous clinical studies. 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. Indeed, as Dr. Bergstrom correctly
`
`notes (EX. 1013 11 33), large numbers of patients are needed to establish a
`
`pharmacokinetic-pharmacodynamic link.
`
`I agree.
`
`C)
`
`Targeted Blood Plasma Drug Concentrations During Therapy
`
`52.
`
`If a relationship between plasma concentrations and response—
`
`efficacy and/or adverse effects—can be established for a drug, that may allow for
`
`the development of a strategy involving achieving and maintaining a target
`
`concentration or a target range of concentrations for individual patients.
`
`53.
`
`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 dosing regimen for patients.
`
`54.
`
`It may be important to monitor plasma concentrations in individual
`
`patients during therapy if one wishes to ensure that those levels are within the
`
`16
`
`AstraZeneca Exhibit 2003 p. 19
`
`

`

`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. In any event, clinicians
`
`who have the responsibility for the care of patients in oncology are typically well
`
`informed about monitoring patient response, including assessing therapeutic
`
`efficacy, and the incidence of troublesome drug related side effects, and making a
`
`change in his or her drug therapy as the situation requires it.
`
`55.
`
`The minimum effective plasma concentration of a drug (MEC), may
`
`also be considered to be a minimum target concentration for a patient receiving
`
`medication on a multiple dosing regimen. A minimum toxic concentration of a
`
`drug (MTC), if established, would represent the upper end of this target range. The
`
`range of plasma concentrations between the MEC and MTC is referred to as the
`
`“therapeutic window.” Patient factors, such as differences in receptor density,
`
`protein binding, and disease state, may contribute to variability in this range in
`
`some patients. Nevertheless, therapeutic windows are considered for many drugs
`
`to represent a range of concentrations within which the likelihood of a desired
`
`clinical effect is relatively high, and that of unacceptable toxicity is relatively low.
`
`56.
`
`It should be noted that response to some drugs changes with plasma
`
`drug concentrations closely in time. For other drugs the response may change
`
`17
`
`AstraZeneca Exhibit 2003 p. 20
`
`

`

`more slowly than the plasma concentrations; this may result from an equilibrium
`
`delay between drug levels in the bloodstream and those at the site of action in the
`
`body, as alluded to above. Such a delay may result in a slow onset of effect, and
`
`may allow the desired response to continue with the same intensity even though the
`
`plasma drug concentrations are decreasing. Whether drug effects are closely
`
`associated in time with plasma drug levels, or exhibit a delay in onset or an
`
`extended duration of action that is unexpected in view of declining plasma levels,
`
`depends on the mechanism of action of the drug. Detailed pharrnacokinetic—
`
`pharmacodynamic modeling studies are often necessary to understand any linkage
`
`between plasma drug concentrations and the response (e.g., the desired therapeutic
`
`effects) observed for a particular drug.
`
`IX) OVERVIEW OF THE PROSECUTION HISTORY
`
`57.
`
`There are a number of inaccuracies in Dr. Bergstrom’s description of
`
`the prosecution history. However, because I do not understand Dr. Bergstrom’s
`
`paragraphs 34-48 and 53-59 to be relevant to his opinions regarding the validity of
`
`the challenged claims, I comment here only with regard to the Declaration Under
`
`37 CPR. § 1.132 of Ronald J. Sawchuk, dated January 13, 2012 and filed during
`
`the prosecution of the ’680 patent (Ex. 1019) (the “Sawchuk Declaration”), which I
`
`authored. The Sawchuk Declaration is discussed below.
`
`A)
`
`The Sawchuk Declaration Describes Numerous Differences That
`Exist Between McLeskey and the Patent Claims
`
`18
`
`AstraZeneca Exhibit 2003 p. 21
`
`

`

`58. With respect to the information in McLeskey regarding the castor oil
`
`fulvestrant composition, the Sawchuk Declaration explains that “[i]n a liquid
`
`composition, when a solute or cosolvent is a liquid, it is often convenient to
`
`express its concentration as a volume percent, i.e., % v/v.” Ex. 1019 at 11 17.
`
`Citing numerous prior art examples where that is the case, i.e., prior art references
`
`that express the concentration of liquid solutes or cosolvents in liquid compositions
`
`as volume percent (% v/v), the Sawchuk Declaration states my belief that “one of
`
`ordinary skill in the art would have concluded the McLeskey castor oil
`
`composition was described in volume/volume units (% v/v).” Ex. 1019 at 11 17. In
`
`reaching that opinion in the Sawchuk Declaration, I did not consider the patents
`
`because they are not part of the state of the art “prior to January 10, 2000.” Ex.
`
`1019 at 11 15.
`
`I do, however, note that Dr. McLeskey herself testified she believed
`
`the McLeskey castor oil composition described to be in volume/volume units (%
`
`v/v). Ex. 2043 at 11 8.
`
`59. With respect to Dr. Bergstrom’s paragraphs 49-52, I understand these
`
`paragraphs to be related to Dr. Bergstrom’s opinions regarding validity of the
`
`challenged claims. Accordingly I address these points below in my analysis of
`
`why, in my opinion, the claimed therapeutically significant blood plasma
`
`fulvestrant concentrations are novel and why one of ordinary skill in the art would
`
`neither combine Howell and McLeskey, nor be motivated to find or reasonably
`
`l9
`
`AstraZeneca Exhibit 2003 p. 22
`
`

`

`expect any formulation described in McLeskey to exhibit the same or similar
`
`pharmacokinetics described in Howell.
`
`B)
`
`The Gellert Declaration and the Sawchuk Declaration Are
`Consistent and Both Support the Patentability 0f the Challenged
`Claims
`
`60.
`
`In paragraphs 53-57, Dr. Bergstrom attempts to re-write certain facts
`
`of the prosecution history of the ’680 patent prosecution to suggest that the
`
`Sawchuk Declaration was inconsistent with the Declaration Under 37 CPR. §
`
`1.132 of Paul Richard Gellert, dated August 8, 2008 and filed during the
`
`prosecution of the ’ 160 patent (Ex. 1020) (the “Gellert Declaration”).1 Contrary to
`
`Dr. Bergstrom’s assertions, the facts of the prosecution history clearly establish the
`
`Gellert and Sawchuk declarations are consistent, and both support the patentability
`
`of the challenged claims. Based on the clear facts in the prosecution history, Dr.
`
`Bergstrom is incorrect.
`
`61.
`
`First, the Gellert and Sawchuk declarations are written from different
`
`perspectives. Unlike the Gellert Declaration, the Sawchuk Declaration is written
`
`from the perspective of one of ordinary skill in the art without the benefit of the
`
`inventors’ confidential research. The purpose of the Sawchuk Declaration was to
`
`“explain how a person of ordinary skill in that art at that time [i.e., prior to January
`
`
`
`1 The Gellert Declaration was attached as an exhibit to the Sawchuk Declaration.
`
`See Ex. 1019 at page 27.
`
`20
`
`AstraZeneca Exhibit 2003 p. 23
`
`

`

`10, 2000] would have understood the references cited in the Office Action and how
`
`such a person would have interpreted certain experimental results related to various
`
`fulvestrant formulations.” Ex. 1019 at 11 15.
`
`62.
`
`On the other hand, the Gellert Declaration, which i

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