throbber
UNITED STATES PATENT AND TRADEMARK OFFICE
`
`BEFORE THE PATENT TRIAL AND APPEAL BOARD
`
`_________________
`
`COMPLEX INNOVATIONS, LLC,
`Petitioner,
`
`v.
`
`AMGEN INCORPORATED
`
`Patent Owner
`
`_________________
`
`U.S. Patent 7,829,595
`
`_________________
`
`
`
`DECLARATION OF WALTER G. CHAMBLISS, Ph.D.
`
`
`1
`
`
`
`
`
`
`
`EX 1021
`IPR of U.S. Pat. No. 7,829,595
`
`

`
`
`
`I, Walter G. Chambliss, declare as follows:
`
`
`1.
`
`I have been retained by Hill, Kertscher, & Wharton, LLP, which
`
`represents Complex Innovations, LLC, in connection with a petition for inter
`
`partes review of U.S. Patent No. 7,829,595, titled Rapid Dissolution Formulation
`
`of a Calcium Receptor-Active Compound (“595 Patent”). I understand that the 595
`
`Patent is currently assigned to Amgen, Incorporated.
`
`
`
`SCOPE OF ANALYSIS
`
`2.
`
`I have reviewed and am familiar with the 595 Patent, which issued to
`
`Lawrence, et al. on November 9, 2010. I understand that the 595 Patent includes
`
`25 claims. I also understand that the Petition for inter partes review that
`
`accompanies this Declaration seeks to cancel claims 1-25 of the 595 Patent. My
`
`analysis and opinions will focus on all challenged claims 1-25.
`
`3. My analysis assumes that the time of invention is September 12, 2003,
`
`which is, in this case, the filing date for provisional application 60/502,219.
`
`4.
`
`I have reviewed and am familiar with various references, written
`
`materials, and literature, which are itemized below:
`
`a) Ex. 1001 U.S. Patent No. 7,829,595 to Lawrence, et al. (“595
`
`Patent”)
`
`b) Ex. 1002
`
`File History to the 595 Patent (“File History”)
`
`
`
`2
`
`

`
`
`
`
`
`c) Ex. 1003 U.S. Patent No. 6,211,244 to Van Wagenen, et al.
`
`(“Van Wagenen”)
`
`d) Ex. 1004 U.S. Patent No. 5,162,117 to Stupak, et al. (“Stupak”)
`
`e) Ex. 1005
`
`European Patent Application No. 1 321 142 A1 by
`
`Vitzling, et al. (“Vitzling”)
`
`f) Ex. 1006 U.S. Patent No. 5,879,706 to Carter, et al. (“Carter”)
`
`g) Ex. 1007 Canadian Patent Application No. 2,004,565 by Chang, et
`
`al. (“Chang”)
`
`h) Ex. 1008 U.S. Patent No. 8,703,196 to Babcock, et al. (“Babcock”)
`
`i) Ex. 1009 U.S. Patent No. 6,733,780 to Tyler, et al. (“Tyler”)
`
`j) Ex. 1010 U.S. Patent No. 4,931,286 to Johnson, et al. (“Johnson”)
`
`k) Ex. 1011
`
`Excerpts from The Pharmaceutical Codex: Principles and
`
`Practice of Pharmaceutics (12th ed.) (1994) (“Pharmaceutical Codex”)
`
`l) Ex. 1012
`
` Excerpts from the Handbook of Pharmaceutical
`
`Excipients (3rd ed.) (2000) (“HPE”)
`
`m) Ex. 1013
`
`Excerpts from Howard C. Ansel, et al., Pharmaceutical
`
`Dosage Forms and Drug Delivery Systems (7th ed.) (1999) (“Ansel”)
`
`n) Ex. 1014
`
`Excerpts from Herbert A. Lieberman, Leon Lachman,
`
`Joseph B. Schwartz (eds.), Pharmaceutical Dosage Forms: Tablets
`
`(2nd ed.) (1989) Vol. 1 (“Lieberman I”)
`
`3
`
`

`
`
`
`o) Ex. 1015
`
`Excerpts from Herbert A. Lieberman, Leon Lachman,
`
`Joseph B. Schwartz (eds.), Pharmaceutical Dosage Forms: Tablets
`
`(2nd ed.) (1989) Vol. 2 (“Lieberman II”)
`
`p) Ex. 1016
`
`Particle Size – U.S. Sieve Series and Tyler Mesh Series
`
`Equivalents (2002; updated 2013), found at
`
`www.azom.com/article.aspx?ArticleID=1417, last accessed August 7,
`
`2015 (“Particle Size”)
`
`q) Ex. 1017 New Agent Reduces PTH Levels in Hemodialysis Patients
`
`With Secondary Hyperparathyroidism, FORMULARY (April 2003)
`
`Vol. 38, p. 197 (“Formulary”)
`
`r) Ex. 1018
`
`J.C. Chaumeil, Micronization: A Method of Improving
`
`the Bioavailability of Poorly Soluble Drugs, METH. FIND. EXP. CLIN.
`
`PHARMACOL. (1998), pp. 211-215 (“Chaumeil”)
`
`s) Ex. 1019
`
`Excerpt from Sir Colin Dollery (ed.), Therapeutic Drugs,
`
`(1991) Vol. 2 (“Dollery”)
`
`t) Ex. 1020 Gordon T. McInnes, et al., Effect of Micronization on the
`
`Bioavailability and Pharmacologic Activity of Spironolactone, J.
`
`CLIN. PHARMACOL. 22 (1982), pp. 410-417 (“McInnes”)
`
`5.
`
`I have been asked to consider how a person of ordinary skill in the art
`
`(“POSITA”) would have understood the claims subject to inter partes review in
`
`
`
`4
`
`

`
`
`
`light of the disclosure of the 595 Patent. I also have been asked how a POSITA
`
`would have understood and applied various references including the Van Wagenen
`
`Patent and the HPE, and whether various other references support them, including
`
`the Stupak, Carter, Babcock, Tyler, and Johnson Patents, the Vitzling and Chang
`
`Patent Applications, and certain material from Ansel, the Pharmaceutical Codex,
`
`Lieberman I, and Lieberman II.
`
`6.
`
`I am being compensated at my standard hourly rate of $800 dollars
`
`per hour for regular consulting and $1000 per hour for live testimony. My
`
`compensation is not dependent on the outcome of this inter partes review and in no
`
`way affects the substance of my testimony in this matter.
`
`
`
`QUALIFICATIONS AND EXPERTISE
`
`7. My resume/curriculum vitae is attached to this declaration as
`
`Exhibit A.
`
`8.
`
`For over thirty-eight (38) years, I have been active in the field of
`
`pharmacy, with over thirty (30) active years in pharmaceutical drug development.
`
`9.
`
`I hold a doctoral degree (Ph.D.) in Pharmaceutics granted by the
`
`University of Mississippi in 1982, as well as a Master of Science in Pharmaceutics
`
`and Bachelor of Science in Pharmacy, granted by the University of Mississippi in
`
`1980 and 1977, respectively.
`
`
`
`5
`
`

`
`
`
`10.
`
`I am currently the Director of Technology Management for the
`
`University of Mississippi, a Research Professor with the Research Institute of
`
`Pharmaceutical Sciences, and a Pharmaceutics Professor at the University of
`
`Mississippi, where I have been a faculty member since 1999. My recent teaching
`
`responsibilities include lectures in undergraduate, graduate and post-graduate
`
`courses related to pharmaceutical formulations, pharmaceutical manufacturing, and
`
`regulatory requirements for the development and manufacture of pharmaceutical
`
`products.
`
`11.
`
`In addition to teaching, over the last eleven (11) years I have been
`
`responsible for managing late stage pharmaceutical development projects, as well
`
`as managing intellectual property and licensing for the University.
`
`12. Prior to joining the faculty at the University of Mississippi, I spent
`
`eleven (11) years with Schering-Plough Healthcare Products, where I was involved
`
`in the launch of approximately thirty (30) new or improved products per year. I
`
`also served as the Vice President of Research and Development from 1993-1998.
`
`
`
`A PERSON OF ORDINARY SKILL IN THE ART
`
`13.
`
`I have been advised and understand that a person having ordinary skill
`
`in the art (“POSITA”) is presumed to be aware of all pertinent art, thinks along
`
`conventional wisdom in the art, and is a person of ordinary creativity. With this
`
`
`
`6
`
`

`
`
`
`understanding, a POSITA at the time of the invention claimed in the 595 Patent is
`
`a person holding a Bachelor of Science degree (or an equivalent) in chemistry,
`
`pharmacy or a related field with at least two years of relevant experience in
`
`pharmaceutical drug development, including formulation work.
`
`
`
`MY UNDERSTANDING OF CLAIM CONSTRUCTION
`
`14.
`
`I have been advised and understand that the claims are to be given
`
`their broadest reasonable construction in light of the specification as it would be
`
`read by a POSITA at the time of invention. I believe that all of the terms of the
`
`claims I have examined have plain and ordinary meanings to a POSITA and no
`
`construction of these terms is necessary.
`
`
`
`MY UNDERSTANDING OF OBVIOUSNESS
`
`15.
`
`I have been advised and understand that a claimed invention is
`
`unpatentable if the differences between the invention and the prior art are such that
`
`the subject matter as a whole would have been obvious to a POSITA at the time
`
`the invention was made to which the subject matter pertains.
`
`16.
`
`It is my understanding that obviousness is a question of law based on
`
`underlying factual findings: (1) the scope and content of the prior art; (2) the
`
`
`
`7
`
`

`
`
`
`differences between the claims and the prior art; (3) the level of skill in the art; and
`
`(4) objective considerations of nonobviousness.
`
`17.
`
`I understand that for one or more references to render the claimed
`
`invention obvious, a POSITA must have a sufficient reason to combine the
`
`teachings of the references to arrive at the claims. I further understand that a basis
`
`to combine teachings from the references need not be stated expressly in any prior
`
`art reference. However, there must be an articulated reason with rational
`
`underpinnings to support a motivation to combine teachings.
`
`18.
`
`I understand that, when considering whether a patent claim is obvious,
`
`a POSITA should consider whether a teaching, suggestion, or motivation to
`
`combine the references exists so as to avoid impermissibly applying hindsight.
`
`
`
`PHARMACEUTICAL DRUG FORMULATIONS, GENERALLY
`
`19. The development of a pharmaceutical composition at the time the
`
`subject patent was filed, as well as in the present time, involved combining a
`
`chemical compound, which has promising therapeutic activity, known in the art as
`
`the drug substance or an active pharmaceutical ingredient (“API”), with one or
`
`more pharmaceutically acceptable inactive ingredients (known in the art as
`
`“excipients,” “adjuvants,” “carriers,” or “additives”) to develop a pharmaceutical
`
`dosage form such as a tablet or a capsule for oral administration. I will use the
`
`
`
`8
`
`

`
`
`
`present tense to describe the basic steps followed to develop a pharmaceutical
`
`formulation since the same steps are used today and also were used at the time the
`
`595 Patent was filed.
`
`20. The goal in developing an oral dosage form is to design a stable
`
`formulation that can be manufactured at a commercial scale, can be self-
`
`administered by the patient, and will release the API in the gastrointestinal tract at
`
`the desired rate. The first step in developing a pharmaceutical formulation is to
`
`characterize the API in a series of routine and well-known studies known in the art
`
`as preformulation studies. (Pharmaceutical Codex at pp. 178-197). I was taught
`
`how to conduct preformulation studies in graduate school in the late 1970s to early
`
`1980s and conducted or supervised the conduct of preformulation studies in the
`
`1980s and 1990s at three different pharmaceutical companies. The overall purpose
`
`of preformulation studies is to determine if there are potential formulation
`
`development issues that a formulator would need to address when developing a
`
`pharmaceutically acceptable dosage form containing the API.
`
`21. Preformulation studies to characterize the API include: determining
`
`the various salt forms of the API; determining the aqueous solubility of the API at
`
`various pH levels, determining the particle size of the API; determining the
`
`morphology of the API (e.g., degree of crystallinity and the shape of the crystals);
`
`
`
`9
`
`

`
`
`
`and determining the stability of the API in the presence of moisture, light, elevated
`
`temperature, and oxygen.
`
`22. The majority of APIs are crystalline, however APIs often contain
`
`some amorphous particles. (Pharmaceutical Codex at p. 179). Common crystal
`
`shapes include cubic and needle shaped. (Id. at p. 180). The optimal shape of the
`
`crystals is determined during preformulation studies and the shape can be altered
`
`through commonly known processing techniques. (Id. at pp. 179-180). It is
`
`commonly known that needle shaped particles will have poor flow properties. (Id.
`
`at p. 180).
`
`23. Based on the data obtained in the preformulation studies, a series of
`
`prototype formulations are made and evaluated for various properties such as
`
`physical and chemical stability, manufacturability, in vitro dissolution, in vitro
`
`disintegration, and in vivo oral bioavailability.
`
`24. There are many different, well-known chemical compounds used as
`
`excipients in pharmaceutical formulations. A POSITA would be very familiar with
`
`the standard treatises that discuss excipients commonly used in pharmaceutical
`
`formulations. Based on my experience, the Handbook of Pharmaceutical
`
`Excipients (the “HPE”) has been the most commonly used reference book on
`
`pharmaceutical excipients for over twenty (20) years. I served on the International
`
`Steering Committee for the 2nd, 3rd and 4th Editions of the HPE and wrote
`
`
`
`10
`
`

`
`
`
`monographs for excipients included in several editions. A POSITA looking for
`
`information on excipients commonly used in tablet formulations would refer to the
`
`HPE for information on the chemistry, physical properties, and applications in
`
`pharmaceutical formulations of various excipients.
`
`25. Excipients are generally classified into categories based on
`
`functionality in the formulation. (See, e.g., Ansel, pp. 87-92; Lieberman I at pp. 91-
`
`121). A tablet formulation, for example, typically would contain as a minimum, the
`
`API, a binder/diluent (also known in the art as a filler) to add bulk to the tablet
`
`such that a patient can hold the tablet in his/her hand, a disintegrant to cause the
`
`tablet to break apart after it is swallowed, and a lubricant to keep the powder blend
`
`from sticking to parts of the tablet press during the compression process.
`
`26. The influence of excipients commonly used in pharmaceutical
`
`formulations on the stability of the API is determined in what are known as
`
`excipient-compatibility studies. (Pharmaceutical Codex, pp. 192-196). From
`
`excipient-compatibility studies, a formulator would learn which excipients could
`
`be used in the solid oral dosage form and which excipients should be avoided.
`
`Typically the API is mixed with each excipient and the mixture is tested under
`
`accelerated conditions. (Id.). In addition, the formulator selects which of the well-
`
`known, off-the-shelf, excipients to use in a formulation based on whether he/she is
`
`developing an immediate release or a modified release tablet or capsule. In the case
`
`
`
`11
`
`

`
`
`
`of an immediate release dosage form a person of ordinary skill can simply select
`
`which excipients to use in prototype formulations based on the results of excipient-
`
`compatibility studies and his/her general knowledge of the functions of excipients
`
`commonly used in immediate release tablets and capsules, such as fillers,
`
`disintegrants and lubricants that do not function to slow down the release of the
`
`API.
`
`27. Prototype formulations will be developed based on the information
`
`learned during preformulation studies. In the case of solid oral dosage forms, such
`
`as tablets and capsules, the amount of each excipient to add to a formulation is
`
`based on considerations such as flowability of the powder, compressibility of the
`
`powder, moisture content of the powder blend and the final dosage form,
`
`disintegration time for the dosage form, dissolution rate of the API from the dosage
`
`form, and stability of the dosage form. (Id. at p. 196).
`
`28. The particle size of the API can affect the dissolution rate of the API
`
`from the dosage form, which likewise impacts the rate and extent at which the API
`
`will be absorbed in the body, the stability of the API, the manufacturability of the
`
`formulation, and the homogeneity of the dosage form. (See, e.g., Lieberman I at
`
`pp. 5-6; Pharmaceutical Codex at p. 181). A desired particle size range for the API
`
`is established during preformulation studies and through routine optimization.
`
`Problems can arise if the mean particle size of the API is too small such as a
`
`
`
`12
`
`

`
`
`
`buildup of static charge, poor mixing and reduced stability. (Pharmaceutical Codex
`
`at pp. 181-184; Lieberman I at p. 5-6). Likewise, problems can arise if the mean
`
`particle size of the API is too large such as poor mixing, variable content
`
`uniformity, reduced rate of dissolution and poor in vivo bioavailability. (Id.).
`
`Generally, a formulator will be motivated to reduce the mean particle size of an
`
`API that has particles greater than approximately 100 microns by grinding the API
`
`using standard milling equipment (also known in the art as “milling”). (Lieberman
`
`I at pp. 5-6). The particle size distribution of an API is easily determined and
`
`typically the median size and some indication of the distribution around the median
`
`are determined. (Pharmaceutical Codex at pp. 181-182; see, e.g., Lieberman II).
`
`29. A common way of manufacturing solid oral dosage forms is by
`
`mixing the API with the excipients and filling the powder blend into empty capsule
`
`shells, or compressing the powder blend into a tablet. This process is referred to in
`
`the art as direct compression in the case of a tablet and dry mixing in the case of a
`
`capsule.
`
`30. Another common way of manufacturing a solid oral dosage forms is
`
`called “wet granulation”. In this case, the powder blend, containing a binder (an
`
`excipient that acts like a glue to hold the powder particles together) is wetted,
`
`typically with water, dried and milled to the desired granule size to ensure good
`
`flowability on a high speed tablet press or encapsulation machine. (See, e.g., Ansel
`
`
`
`13
`
`

`
`
`
`at pp. 209-211). In the case of tablets, the desired size of granules depends on the
`
`size of the tablet punches that will be used to compress the tablet. (Id. at p. 211
`
`(“In general, the smaller the tablet to be produced, the smaller are the granules
`
`used”.)) Typically, granules resulting from a wet granulation process are milled to
`
`have a mean particle size between 12 and 20 Tyler mesh, which equates to between
`
`about 80 to about 140 microns. (Id. at p. 211; Particle Size at pp. 1-2).
`
`31. Tablets are commonly coated with a polymer (known in the art as a
`
`“film coat”) or sugar based coating. The coating can be clear or a pigment can be
`
`added to impart color. Coatings are added to, among other purposes, protect tablets
`
`from a humid environment, mask the taste of a bad tasting API, and make the
`
`tablet easier to swallow. (Ansel at pp. 219-227). A film coat also can be used to
`
`modify the release of the API from the dosage form. (Id.)
`
`
`
`ANALYSIS OF THE 595 PATENT (Ex. 1001)
`
`32. The 595 Patent relates to the development of pharmaceutical
`
`compositions including tablets used in the treatment of, among other things,
`
`secondary hyperparathyroidism commonly associated with chronic kidney disease.
`
`(595 Patent, col. 4, lines 16-21; Formulary at p.197). To be more specific, the
`
`chemistry described in the 595 Patent pertains to a pharmaceutical composition
`
`
`
`14
`
`

`
`
`
`comprising cinacalcet hydrochloride in combination with excipients commonly
`
`used in tablet formulations. (E.g., id., claim 1 & Examples, col. 11-13).
`
`33. Hyperparathyroidism results from impaired calcium homeostasis.
`
`(Van Wagenen, col. 6, lines 9-10). It is indicated by high levels of the parathyroid
`
`hormone (“PTH”) in the bloodstream. (Id., col. 16, lines 66-67). Further, increased
`
`levels of free calcium in the bloodstream is also associated with secondary
`
`hyperparathyroidism. (Id., col 17, line 44 – col. 18, line 15). Cinacalcet
`
`hydrochloride is a calcium receptor-active compound (595 Patent, col. 1, lines 7-
`
`10) that can simultaneously reduce PTH and control calcium-phosphorus product
`
`in the bloodstream (Formulary at p.197).
`
`34. Although the 595 Patent was filed on September 10, 2004, as noted
`
`above, my analysis assumes that the time of invention is September 12, 2003 (the
`
`filing date for provisional application 60/502,219).
`
`35. The 595 Patent admits that cinacalcet hydrochloride was known in the
`
`art prior to the filing of the 595 Patent. (595 Patent, col. 1, lines 7-10 & Certificate
`
`of Correction at p.10) (“One example of a calcium receptor-active compound is
`
`cinacalcet HCl, which is described, for example, in U.S. Pat. No. 6,211,244 [Van
`
`Wagenen]”). Indeed, a number of patents filed prior to the 595 Patent cover
`
`cinacalcet hydrochloride, as demonstrated by their listing in the Food and Drug
`
`Administration’s Orange Book
`
`
`
`15
`
`

`
`
`
`(see http://www.accessdata.fda.gov/scripts/cder/ob/docs/patexclnew.cfm?Appl_No
`
`=021688&Product_No=001&table1=OB_Rx, last accessed August 12, 2015) and
`
`reference to them in the 595 Patent specification (col. 2, lines 18-28).
`
`36. Further, Van Wagenen teaches pharmaceutical compositions
`
`containing an API and an acceptable carrier. (Van Wagenen, col. 5, lines 26-35).
`
`37. Additionally, the treatment of hyperparathyroidism with a
`
`pharmaceutical composition comprising a therapeutically effective amount of an
`
`inorganic ion receptor-modulating agent, such as cinacalcet hydrochloride, was
`
`well known to a POSITA prior to the time of invention. (See, e.g., Van Wagenen,
`
`col. 5, line 48 – col. 6, line 18.)
`
`38. As discussed above and further below, at the time of the invention, the
`
`use of various excipients in the formulation of pharmaceutical products, including
`
`tablets, was well known. The 595 Patent uses then well-known formulation
`
`techniques to the formulation of the cinacalcet hydrochloride composition of
`
`claim 1.
`
`39. The specification of the 595 Patent focuses on an improved
`
`dissolution profile for a cinacalcet hydrochloride composition consisting of
`
`granules. (See, e.g., 595 Patent, col. 9, lines 22-48). According to the specification,
`
`the invention relates to “a pharmaceutical composition comprising a
`
`therapeutically effective amount of a calcium receptor-active compound and at
`
`
`
`16
`
`

`
`
`
`least one pharmaceutically acceptable excipient, wherein the composition has a
`
`controlled dissolution profile.” (Id., abstract). However, the claims of the 595
`
`Patent do not include any reference to a dissolution profile, improved or otherwise.
`
`40. The file history for the application that resulted in the ‘595 Patent
`
`supports my opinion that the claims discussed in this declaration are obvious in
`
`light of the general knowledge of a POSITA as bolstered by numerous prior art
`
`references. To the extent applicants argue unexpected or surprising results, I
`
`disagree that the specification or the file history reflects any unexpected or
`
`surprising results. During prosecution, applicants asserted to the USPTO that “the
`
`poor water solubility of cinacalcet hydrochloride reduces the options for
`
`formulating the compound for effective delivery and bioavailability.” (File History
`
`- Amendment and Reply Under 37 CFR 1.116, July 14, 2008 at p. 10). Applicants
`
`further asserted to the USPTO that “the skilled person would not harbor an
`
`expectation of formulating cinacalcet hydrochloride in an acceptably soluble and
`
`readily bioavailable composition with just any given combination of excipients”
`
`because the poor water solubility of this cinacalcet hydrochloride allegedly “limits
`
`options for formulations and can adversely impact the bioavailability of cinacalcet
`
`hydrochloride”). (Id. - Supplemental Amendment and Reply Under 37 CFR 1.111,
`
`December 23, 2008 at p. 8).
`
`
`
`17
`
`

`
`
`
`41. However, what the Applicants did not tell the USPTO was that at the
`
`time, formulation techniques for APIs with low solubility were well known. Based
`
`on my experience, and consistent with the literature, some APIs are highly water
`
`soluble, but the water solubility of many APIs is low, especially for APIs
`
`developed since the late 1990s. (See, e.g., Chaumeil at p. 211). It was well known
`
`in the art how to develop successful pharmaceutical formulations containing an
`
`API with poor water solubility. For example, Chaumeil teaches reducing the
`
`particle size of several APIs with poor water solubility in order to increase oral
`
`bioavailability. (See generally Chaumeil).
`
`42. Further, I have developed pharmaceutical formulations for a number
`
`of APIs that have poor water solubility, including spironolactone, which is water
`
`insoluble. (Dollery at S85). It has been known since the early 1980s that reducing
`
`the particle size of spironolactone will improve oral bioavailability. (See, e.g.,
`
`McInnes at pp. 410-417 (teaching the reduction of median particle size of
`
`spironolactone from 78.8 μm to 2.21 μm significantly increases oral bioavailability
`
`without any changes to the excipients contained in the tablet formulation)).
`
`
`
`ANALYSIS OF THE 244 PATENT (VAN WAGENEN – EX. 1003)
`
`43.
`
`I have reviewed and am familiar with U.S. Patent. No. 6,211,244
`
`issued to Van Wagenen, et al. (Ex. 1003 – Van Wagenen).
`
`
`
`18
`
`

`
`
`
`44. Van Wagenen (“Calcium Receptor-Active Compounds”) is a U.S.
`
`Patent disclosing calcium receptor-active compounds, including cinacalcet
`
`hydrochloride. (Id., col. 1, lines 1-9).
`
`45. Among the topics covered in Van Wagenen is the treatment of
`
`hyperparathyroidism by administering a pharmaceutical composition comprising a
`
`therapeutically effective amount of an inorganic ion receptor-modulating agent.
`
`(Id., col. 5, line 48 – col. 6, line 24; col. 17, line 44 – col. 18, line 15).
`
`46. Van Wagenen also teaches that “suitable dosage ranges, formulations,
`
`and dosage forms” can be “determined by one skilled in art based on the teachings
`
`provided in the application.” (Id., col. 44, lines 39-45).
`
`
`
`ANALYSIS OF THE HANDBOOK OF PHARMACEUTICAL EXCIPIENTS
`(“HPE”) (3rd ED.) (EX. 1012)
`
`47.
`
`In selecting particular excipients for use in a pharmaceutical
`
`composition containing cinacalcet hydrochloride, as discussed above, one skilled
`
`in the art would be aware of the HPE as a resource providing information on the
`
`uses of a variety of pharmaceutical excipients in composition formulation. The 3rd
`
`Edition of the HPE disclosed numerous excipients for use in pharmaceutical
`
`formulations in 2000. As discussed above, I was heavily involved in the editing of
`
`this HPE. The teachings of HPE are intended to be applied to APIs in general
`
`rather than being limited to specific APIs.
`19
`
`
`
`

`
`
`
`48. Among the excipients covered in the HPE are microcrystalline
`
`cellulose, povidone, starch, crospovidone, colloidal silicon dioxide, and
`
`magnesium stearate. (See generally HPE).
`
`
`
`ANALYSIS OF ANSEL (EX. 1013)
`
`49. Ansel, a well-known pharmaceutical treatise, teaches among other
`
`things the development of general types of pharmaceutical compositions (e.g.,
`
`granules, tablets, capsules and powders), the importance of particle size of the API
`
`and/or granulations, and the common types of excipients used in pharmaceutical
`
`compositions such as tablets (e.g., lubricants, binders, fillers and coating
`
`materials). (See generally Ansel).
`
`50. Among the excipients covered in Ansel are microcrystalline cellulose,
`
`povidone, starch, crospovidone, colloidal silicon dioxide, and magnesium stearate.
`
`(See generally Ansel).
`
`
`
`ANALYSIS OF THE 117 PATENT (STUPAK – EX. 1004)
`
`51.
`
`I have reviewed and am familiar with U.S. Pat. No. 5,162,117 to
`
`Stupak, et al. (Ex. 1004 – Stupak).
`
`52. Stupak is a U.S. Patent disclosing a controlled release flutamide
`
`composition. (Id., col. 1, lines 1-3).
`
`
`
`20
`
`

`
`
`
`53. Among other topics, Stupak discloses the use of starch in a
`
`pharmaceutical composition in the range of 15% to 35%. (Id., col. 2, lines 16-26).
`
`
`
`ANALYSIS OF THE 142 APPLICATION (VITZLING – EX. 1005)
`
`54.
`
`I have reviewed and am familiar with European Patent Application
`
`No. 1 321 142 A1 by Vitzling, et al. (Ex. 1005 – Vitzling).
`
`55. Vitzling is a European Patent Application for a solid pharmaceutical
`
`composition for oral administration of Tegaserod. (Id. at p.1).
`
`56. Among other topics, Vitzling discloses a pharmaceutical composition
`
`including 15% or less of a disintegrant, including, but not limited to crospovidone.
`
`(Id. at p. 2, ¶¶ 9 & 11).
`
`
`
`ANALYSIS OF THE 706 PATENT (CARTER – EX. 1006)
`
`57.
`
`I have reviewed and am familiar with U.S. Patent No. 5,879,706 to
`
`Carter, et al. (Ex. 1006 – Carter).
`
`58. Carter is a U.S. Patent for Valaciclovir tablets containing colloidal
`
`silicon dioxide. (Id., col. 1, lines 1-2).
`
`59. Among other topics, Carter discloses the use of colloidal silicon
`
`dioxide in the range of 0.05% to 3%. (Id., abstract; col. 2, lines 27-61).
`
`
`
`
`
`21
`
`

`
`
`
`ANALYSIS OF THE 565 APPLICATION (CHANG – EX. 1007)
`
`60.
`
`I have reviewed and am familiar with Canadian Patent Application
`
`No. 2,004,565 by Chang, et al. (Ex. 1007 – Chang).
`
`61. Chang is a Canadian Patent Application for a sustained release
`
`diltiazem formulation. (Id. at p.1).
`
`62. Among other topics, Chang discloses the use of magnesium stearate in
`
`the range of 0% to 2%. (Id. at p.9).
`
`
`
`ANALYSIS OF THE 196 PATENT (BABCOCK – EX. 1008)
`
`63.
`
`I have reviewed and am familiar with U.S. Pat. No. 8,703,196 to
`
`Babcock, et al. (Ex. 1008 – Babcock).
`
`64. Babcock is a U.S. Patent disclosing pharmaceutical compositions of
`
`dispersions of amorphous drugs mixed with polymers. (Id., col. 1, lines 1-3).
`
`65. Among other topics, Babcock discloses pharmaceutical compositions
`
`comprising matrix drug particles. (Id., col. 99, lines 58-61).
`
`
`
`ANALYSIS OF THE 780 PATENT (TYLER – EX. 1009)
`
`66.
`
`I have reviewed and am familiar with U.S. Patent No. 6,733,780 to
`
`Tyler, et al. (Ex. 1009 – Tyler).
`
`
`
`22
`
`

`
`
`
`67. Among other teachings, Tyler discloses coating compositions ranging
`
`from about 4% to about 6%. (Id., col. 3, lines 42-54).
`
`
`
`ANALYSIS OF THE 286 PATENT (JOHNSON – EX. 1010)
`
`68.
`
`I have reviewed and am familiar with U.S. Patent No. 4,931,286 to
`
`Johnson, et al. (Ex. 1010 – Johnson).
`
`69. Among other teachings, Johnson discloses coating compositions
`
`ranging from about 0.5% to about 5%. (Id., claim 3).
`
`
`
`ANALYSIS OF THE PHARMACEUTICAL CODEX (EX. 1011)
`
`70. Among the topics covered by the Pharmaceutical Codex, a well-
`
`known pharmaceutical treatise, is the fact that APIs may be crystalline, amorphous
`
`or a combination thereof; and that the optimal shape of crystalline particles is
`
`determined during preformulation studies. (Pharmaceutical Codex at p.179-180).
`
`
`
`ANALYSIS OF LIEBERMAN I (EX. 1014)
`
`71. Lieberman I, a well-known pharmaceutical treatise, teaches, among
`
`other things, example pharmaceutical compositions containing APIs and
`
`commonly used excipients in typical concentrations, and the importance of the
`
`particle size of the API. (See generally Lieberman I).
`
`
`
`23
`
`

`
`
`
`
`
`ANALYSIS OF LIEBERMAN II (EX. 1015)
`
`72. Lieberman II, a well-known pharmaceutical treatise, teaches, among
`
`other things, the importance of the particle size of the API and the use of a sieve
`
`analysis to determine particle distribution. (See generally Lieberman II).
`
`
`
`THE REFERENCED FORMULATIONS AND DOSAGES IN CLAIMS 1-23
`ARE RENDERED OBVIOUS BY PRIOR ART
`
`73. Each element of claims 1-25 is disclosed, shown, or suggested by Van
`
`Wagenen, the HPE, and the general knowledge of a POSITA, as bolstered by
`
`Stupak, Vitzling, Carter, Chang, Babcock, Tyler, Johnson, the Pharmaceutical
`
`Codex, Ansel, Lieberman I and/or Lieberman II. As discussed above, a POSITA
`
`would be motivated to combine the teachings of Van Wagenen and the HPE in
`
`light of the POSITA’s general knowledge. Presented below is my analysis that
`
`addresses each claim element. A POSITA would have been motivated to combine
`
`the Van Wagenen and HPE references based on his/her general knowledge and
`
`supported by the fact that well-known treatises, such as Ansel, Lieberman I, and
`
`Lieberman II, teach tablet formulations using particle sizes and excipients
`
`commonly used to make tablets including excipients used in the core of the tablet
`
`and excipients used in a standard tablet coating layer. Such a combination would
`
`be the natural result of routine formulation development as described above. In
`24
`
`
`
`

`
`
`
`other words, based on this prior art and general knowledge, a POSITA would
`
`expect that a pharmaceutical composition (e.g., a tablet) containing cinacalcet
`
`hydrochloride, a known API, could be made using commonly used excipients
`
`without undue experimentation.
`
`Claim 1
`
`74. Claim 1 states: “A pharmaceutical composition comprising (a) from
`
`about 10% to about 40% by weight of cinacalcet HCl; (b) from about 40% to about
`
`75% by weight of microcrystalline cellulose; (c) from about 1% to about 5% by
`
`weight of povidone; (d) from about 5% to about 35% by weight of starch; (e) from
`
`about 1% to about 10% by weight of crospovidone; (f) from about 0.05% to about
`
`1.5% by weight of colloidal silicon dioxide; and (g) from about 0.05% to about
`
`1.5% by weight of magnesium stearate; wherein the percentage by weight is
`
`relative to the total weight by composition.” (595 Patent, col. 14, lines 17-31).
`
`75.
`
` Van Wagenen teaches pharmaceutical compositions containing
`
`cinacalcet hydrochloride. (Id., col. 2, lines 18-28). Indeed, the 595 P

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