throbber
Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`UNITED STATES PATENT AND TRADEMARK OFFICE
`
`____________________________________________
`
`BEFORE THE PATENT TRIAL AND APPEAL BOARD
`
`____________________________________________
`
`MYLAN PHARMACEUTICALS, INC.,
`Petitioner,
`
`v.
`
`GENENTECH, INC. AND CITY OF HOPE
`Patent Owners.
`
`____________________________________________
`
`Case IPR2016-00710
`Patent 6,331,415
`
`____________________________________________
`
`EXPERT DECLARATION OF JOHN FIDDES, PH.D.
`
`
`
`
`
`
`
`Mylan v. Genentech
`IPR2016-00710
`Genentech Exhibit 2019
`
`

`
`
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`TABLE OF CONTENTS
`
`Page
`
`I. 
`
`INTRODUCTION AND BACKGROUND .................................................... 1 
`
`A.  Qualifications And Experience ............................................................. 2 
`
`B. 
`
`C. 
`
`Compensation ........................................................................................ 4 
`
`Prior Expert Testimony ......................................................................... 4 
`
`II. 
`
`LEGAL PRINCIPLES ON OBVIOUSNESS ................................................. 5 
`
`III.  BACKGROUND OF THE TECHNOLOGY .................................................. 7 
`
`A.  Genes, Proteins And Antibodies ........................................................... 8 
`
`B. 
`
`C. 
`
`Antibody Production Techniques As Of April 1983 .......................... 16 
`
`Use of Recombinant Gene Expression To Produce Proteins .............. 18 
`
`1. 
`
`2. 
`
`Basic principles of recombinant gene expression ..................... 18 
`
`As of April 1983, only a small number of monomeric
`eukaryotic proteins had been produced recombinantly. ........... 20 
`
`D.  As of April 1983, Insulin Was The Only Multimeric Eukaryotic
`Protein Consisting Of Different Polypeptide Chains Which Had
`Been Produced Recombinantly. .......................................................... 29 
`
`E. 
`
`F. 
`
`In May 1981 (When Bujard Was Filed), The Speculative
`Possibility Of Using Recombinant Techniques To Produce
`Antibodies Was Highly Uncertain And Unpredictable ....................... 38 
`
`Research Involving Antibodies And Recombinant Gene
`Expression Between May 1981 And April 1983 Confirmed The
`Uncertainty And Unpredictability Of Whether Antibodies
`Could Be Produced Recombinantly. ................................................... 41 
`
`G.  As Of April 1983, Highly Acclaimed Scientists Were Still
`Uncertain Whether It Was Even Possible To Make Antibodies
`Using Recombinant Techniques. ........................................................ 46 
`
`i
`
`

`
`
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`H.  As Of April 1983, Nobody Had Produced An Antibody
`Recombinantly. .................................................................................... 49 
`
`I. 
`
`As Of April 1983, Nobody Had Produced A Multimeric
`Eukaryotic Protein Encoded By More Than One Gene
`Recombinantly In A Single Host Cell. ................................................ 49 
`
`IV.  THE CLAIMS UNDER CONSIDERATION AND THEIR
`INTERPRETATION ..................................................................................... 53 
`
`A. 
`
`B. 
`
`C. 
`
`The Cabilly ’415 Patent ....................................................................... 53 
`
`Summary Of Contested Claims ........................................................... 54 
`
`The Person Of Ordinary Skill In The Art ............................................ 56 
`
`V.  OPINIONS REGARDING THE ASSERTED PRIOR ART ........................ 57 
`
`A. 
`
`Bujard .................................................................................................. 58 
`
`1.  What is the focus of the Bujard reference? ............................... 58 
`
`2. 
`
`3. 
`
`4. 
`
`5. 
`
`6. 
`
`Do the references to “multimers” and “one or more structural
`genes” in Bujard suggest the use of the co-expression of
`multiple, distinct eukaryotic genes of interest in a single host
`cell? ........................................................................................... 63 
`
`Do the references to multiple “stop codons” in Bujard suggest
`the use of the strong promoter/terminator system for the
`expression of multiple, distinct genes in a single host cell? ..... 73 
`
`Does Bujard “at least suggest the coexpression of the heavy and
`light chains” of an immunoglobulin in a “single host cell,” as
`the Board found? ....................................................................... 75 
`
`Does Bujard “teach away from the production of light chains in
`one culture and heavy chains in another, to be combined
`chemically at a stage after their harvest and isolation,” as Dr.
`Foote argues? ............................................................................ 80 
`
`Does the statement in Bujard that the “proteins may be prepared
`as a single unit or as individual subunits and then joined
`
`ii
`
`

`
`
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`together in appropriate ways” suggest in vivo assembly of a
`multimeric protein encoded by more than one gene in a single
`host cell, as Dr. Foote argues? .................................................. 85 
`
`7. 
`
`Do you agree with the Board’s finding that Bujard is “more
`specific and robust” than the Axel reference? .......................... 87 
`
`B. 
`
`Riggs & Itakura ................................................................................... 88 
`
`1.  What is the focus of the Riggs & Itakura reference? ................ 88 
`
`2. 
`
`Does the Riggs & Itakura reference address the “same problem”
`as Bujard and would the skilled artisan have had “good reason”
`to combine these two references in April 1983? ...................... 90 
`
`3.  Would inferences gleaned from Riggs & Itakura have provided
`the person of ordinary skill with the motivation to “selectively
`apply the teachings of Bujard to the specific production of
`immunoglobulins” by means of co-expressing both the heavy
`and light chain in a single host cell? ......................................... 92 
`
`4. 
`
`Do you agree with Dr. Foote’s opinion and the Board’s
`preliminary finding that Bujard in combination with Riggs &
`Itakura renders the claimed invention of the Cabilly ’415 patent
`obvious? .................................................................................... 99 
`
`C. 
`
`Southern ............................................................................................. 101 
`
`1.  What is the focus of the Southern reference? ......................... 101 
`
`2. 
`
`Do you agree with Dr. Foote’s opinion that the skilled artisan
`would have been motivated to combine Bujard and Southern
`because “both have as a goal the expression of genes of interest
`in a single transformed host cell, whether by using one (Bujard)
`or two (Southern) vectors”? .................................................... 103 
`
`3.  Would inferences gleaned from Southern have provided the
`person of ordinary skill with the motivation to “selectively
`apply the teachings of Bujard to the specific production of
`immunoglobulins” by means of co-expressing both the heavy
`and light chain in a single host cell? ....................................... 105 
`
`iii
`
`

`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`4. 
`
`Do you agree with Dr. Foote’s opinion and the Board’s
`preliminary finding that Bujard in combination with Southern
`renders the claimed invention of the Cabilly ’415 patent
`obvious? .................................................................................. 112 
`
`
`
`
`
`iv
`
`

`
`
`I.
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`INTRODUCTION AND BACKGROUND
`1.
`
`I, John Fiddes, Ph.D., have been retained by counsel for Genentech,
`
`Inc. and City of Hope (collectively, “Patent Owners”) as an expert in this
`
`proceeding.
`
`2.
`
`I understand that, in a September 8, 2016 decision, the Patent Trial and
`
`Appeal Board (the “Board”) instituted inter partes review as to claims 1-4, 11, 12,
`
`14, 18-20, and 33 of U.S. Patent No. 6,331,415 (“the Cabilly ’415 patent”). I
`
`further understand that the references relied upon by the Board in instituting inter
`
`partes review (“IPR”) include the Bujard patent (Ex. 1002), the Riggs & Itakura
`
`paper (Ex. 1003) and the Southern paper (Ex. 1004). (Paper 13 at 12-15.) I
`
`understand that these grounds are the same as those in IPR2015-01624, and that
`
`the Board in this proceeding adopted the reasoning from the institution decision in
`
`IPR2015-01624. (Paper 13 at 3, 14.) As a result, I cite below to the institution
`
`decision from this proceeding (Paper 13) as well as from IPR2015-01624.
`
`(IPR2015-01624, Paper 15.)
`
`3.
`
`I have been asked to review the challenged claims of the Cabilly ’415
`
`patent and the references identified in the petition requesting inter partes review,
`
`and evaluate whether the cited references alone or in combination render the
`
`challenged claims unpatentable. As part of my review I have been asked to
`
`evaluate the prior art and scientific accuracy of the observations that the Board
`
`1
`
`

`
`
`made in the decision instituting inter partes review. I also was asked to evaluate
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`certain statements that Dr. Jefferson Foote made in a declaration (Ex. 1006), which
`
`was submitted in IPR2015-01624, as well as the declaration of Dr. Kathryn
`
`Calame, which adopts the opinions set forth in Dr. Foote’s declaration without
`
`further elaboration. (Ex. 1059, Calame Decl. ¶ 16.) Because Dr. Calame adopted
`
`Dr. Foote’s statements, I cite below to Dr. Foote’s declaration and his opinions.
`
`4. A list of materials I have reviewed in preparation of this Declaration is
`
`attached as Exhibit B. I have also relied upon my scientific knowledge as of April
`
`1983 when the Cabilly ’415 patent was filed.
`
`A. Qualifications And Experience
`5. My background is summarized below and in my curriculum vitae,
`
`which includes a list of my publications and patents and is attached as Exhibit A.
`
`6.
`
`I received a Bachelor of Science degree in Biological Sciences
`
`(Molecular Biology) with First Class Honors from the University of Edinburgh in
`
`1973. In 1977, I received my Ph.D. in Molecular Biology from Kings College,
`
`Cambridge University. My thesis advisor was Dr. Fred Sanger. The title of my
`
`thesis was “The Determination of Nucleotide Sequences in Bacteriophage ΦX174
`
`DNA.”
`
`7.
`
`From 1977 to 1980, I was a Postdoctoral Research Fellow at the
`
`University of California, San Francisco (“UCSF”), in the laboratory of Dr. Howard
`
`2
`
`

`
`
`Goodman, where I worked on the human growth hormone, human chorionic
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`somatomammotropin and human glycoprotein hormone genes.
`
`8. After my post-doc at UCSF, I became a Senior Staff Investigator at
`
`Cold Spring Harbor Laboratory (“CSHL”) in Cold Spring Harbor, New York, a
`
`position I held until January 1983.
`
`9. My research at CSHL focused on the structure, evolution and
`
`expression of the human glycoprotein hormone genes, specifically human
`
`chorionic gonadotropin and human luteinizing hormone, and on methods of
`
`making cDNA libraries suitable for immunological screening of expression
`
`products. I was also an instructor at the CSHL Advanced Cloning Course in the
`
`summers of 1982-1983.
`
`10. Following my academic career, I entered industry and spent over
`
`twenty years in drug discovery and development. In January 1983, just shortly
`
`before the filing date of the Cabilly ’415 patent, I took a position at California
`
`Biotechnology Inc. (later renamed Scios Inc.) in Mountain View, California. The
`
`primary interest of this company was in applying recombinant DNA technologies
`
`to the production of therapeutically useful proteins.
`
`11. Among other things, I was involved in the development of systems for
`
`the production of recombinant forms of basic fibroblast growth factor, and the
`
`3
`
`

`
`
`isolation of cDNA and genomic clones for atrial natriuretic peptide, vascular
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`endothelial growth factor variant and heparin-binding, EGF-like growth factor.
`
`12. My last industry position was at Genencor International Inc. in Palo
`
`Alto, California where I served as Vice President Research, Health Care from 2003
`
`to 2005. Since 2005, I have been an independent consultant on biopharmaceutical
`
`matters for a variety of companies, and for two non-profit organizations, the
`
`California Antiviral Foundation and the Institute for One World Health.
`
`13. Based on my academic and early industry experience, I was well
`
`aware of the birth of recombinant DNA technology and followed the developments
`
`that eventually led to the production of recombinant forms of medically important
`
`proteins. This, in my view, is the art to which the Cabilly ’415 patent pertains, and
`
`I believe I am well-positioned to understand and address the skills and mindset of a
`
`person of ordinary skill in this field circa 1982-1983.
`
`B. Compensation
`14.
`
`I am being compensated at my normal consulting rate for my work,
`
`which is $650 per hour. My compensation is not dependent on and in no way
`
`affects the substance of my statements in this Declaration.
`
`C.
`15.
`
`Prior Expert Testimony
`I provided expert reports and deposition testimony in Bristol-Myers
`
`Squibb Co. v. Genentech, Inc. & City of Hope, 2:13-cv-05400-MRP-JEM (C.D.
`
`4
`
`

`
`
`Cal), Eli Lilly & Co. v. Genentech, Inc., 2:13-cv-07248-MRP-JEM (C.D. Cal.), and
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`Sanofi-Aventis U.S. LLC & Regeneron Pharm., Inc. v. Genentech, Inc. & City of
`
`Hope, 2:15-cv-05685-GW-AGR (C.D. Cal.). I also provided a declaration and
`
`deposition testimony in Sanofi-Aventis U.S. LLC & Regeneron Pharm., Inc. v.
`
`Genentech, Inc. & City of Hope, Case IPR2015-01624 (P.T.A.B.).
`
`II. LEGAL PRINCIPLES ON OBVIOUSNESS
`16.
`I have been informed and understand that in order to invalidate a
`
`patent claim as obvious in the context of an inter partes review, it must be shown
`
`by a preponderance of the evidence that the claim would have been obvious to a
`
`person of ordinary skill at the time the invention was made. The prior art does not
`
`need to render obvious every possible embodiment within the scope of the claim:
`
`the prior art renders the claim obvious if the combined teachings disclose an
`
`embodiment that is within the scope of the claim.
`
`17.
`
`I have been informed and understand that factors relevant to the
`
`determination of obviousness include the scope and content of the prior art, the
`
`level of ordinary skill in the art at the time of the invention, differences between
`
`the claimed invention and the prior art and “secondary considerations” or objective
`
`evidence of non-obviousness.
`
`18.
`
`I have been informed and understand that obviousness can be
`
`established by combining or modifying the teachings of the prior art to produce the
`
`5
`
`

`
`
`claimed invention where there is some teaching, suggestion or motivation to do so;
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`and that a reasonable expectation of success in achieving the subject matter of the
`
`claim at issue must also be shown. Further, I have been informed and understand
`
`that the teaching, suggestion or motivation test is flexible and that an explicit
`
`suggestion to combine the prior art is not necessary – the motivation to combine
`
`may be implicit and may be found in the knowledge of one of ordinary skill in the
`
`art, from the nature of the problem to be solved, market demand or common sense.
`
`19.
`
`I have been informed and understand that a patent claim composed of
`
`several limitations is not obvious merely because each limitation was
`
`independently known in the prior art. Hindsight reasoning is not an appropriate
`
`basis for combining references to form an obviousness combination. I also have
`
`been informed and understand that it can be important to identify a reason that
`
`would have prompted a person of ordinary skill in the relevant field to combine the
`
`limitations in the way the claimed new invention does.
`
`20.
`
`In undertaking an obviousness analysis, I have been informed and
`
`understand that I may take into account the inferences and creative steps that a
`
`person of ordinary skill would have employed in reviewing the prior art at the time
`
`of the invention. If the claimed invention combines elements known in the prior
`
`art and the combination yields results that would have been predictable to a person
`
`6
`
`

`
`
`of ordinary skill at the time of the invention, then this evidence would make it
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`more likely that the claim was obvious.
`
`21.
`
`I have also been informed and understand that obviousness may be
`
`established if the combination of prior art elements was obvious to try, even if no
`
`one attempted the combination. For a combination to be obvious to try, however, a
`
`solution must be among a finite number of identified, predictable solutions. Where
`
`the art is uncertain or unpredictable, a person of ordinary skill in the art will not
`
`have a reasonable expectation of success.
`
`22.
`
`I have been informed and understand that an obviousness analysis
`
`must take into account any “secondary considerations” or, as they are sometimes
`
`called, objective indicia of non-obviousness. These secondary considerations can
`
`include the invention’s commercial success, long-felt but unresolved needs,
`
`licenses showing industry respect, the failure of others, skepticism by experts,
`
`praise by others, teaching away by others, recognition of a problem and copying of
`
`the invention by competitors. Such secondary considerations, when present, offer
`
`objective information as to the state of the art at the time of the invention and
`
`provide a check to hindsight analysis.
`
`III. BACKGROUND OF THE TECHNOLOGY
`23. To place the importance and innovation of the Cabilly ’415 patent into
`
`context, I have been asked to provide some background on the relevant technology.
`
`7
`
`

`
`
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`A. Genes, Proteins And Antibodies
`24. A deoxyribonucleic acid (DNA) molecule encodes the genetic
`
`instructions that a living organism uses for a wide variety of critical functions.
`
`Sequences of DNA nucleotides are organized into discrete structures, called genes,
`
`which a cell’s machinery “reads” to make proteins. Proteins, comprised of a string
`
`of units called amino acids, are biomolecules that perform many of the functions of
`
`cells and organisms.
`
`25. The basic process of making a protein from a gene is called “gene
`
`expression.” First, the cell copies the gene of interest (a DNA sequence) into
`
`messenger ribonucleic acid (mRNA) via a process called transcription. Second,
`
`the mRNA is converted into the corresponding sequence of amino acids (called a
`
`polypeptide) via a process called translation. Finally, the translated polypeptide
`
`undergoes folding and possibly post-translational modifications to assemble as the
`
`active protein structure.
`
`26. This step-wise process is reflected in the following illustration:
`
`8
`
`

`
`
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`
`(Ex. 2081, Bruce Alberts et al., Essential Cell Biology, Chapter 1 (3rd ed. 2009).)
`
`27. Transcription: In transcription, an enzyme in cells called RNA
`
`polymerase synthesizes single-stranded mRNA in a linear fashion from the
`
`doubled-stranded genomic DNA template.
`
`28. Translation: In translation, complex structures in cells called
`
`ribosomes bind to specific sites on the mRNA transcript and translate the mRNA
`
`sequence into a polypeptide chain of amino acids. The mRNA sequence is read
`
`three nucleotides at a time, with each triplet of nucleotides – called a codon –
`
`specifying one amino acid. A start codon initiates translation, and stop codons
`
`signal termination of translation. Codons, therefore, provide the information that
`
`9
`
`

`
`
`dictates the order and arrangement of amino acids in a polypeptide chain, and
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`when the formation of the chain begins and ends.
`
`29. An exemplary coding region (from protein B of bacteriophage G4) is
`
`shown in the table below (see Ex. 2082, G. N. Godson et al., Nucleotide Sequence
`
`of Bacteriophage G4 DNA, Nature 276:236-47 (1978)):
`
`TAC
`Codon
`Amino Acid TYR
`
`GGA
`GLY
`
`TAT
`TYR
`
`TTC
`PHE
`
`TGA
`STOP
`
`TGA
`STOP
`
`30. As seen in this table above, multiple stop codons (written for ease of
`
`reference as DNA as opposed to mRNA) can signal the end of translation of a
`
`single polypeptide. A coding region may end with multiple stop codons to ensure
`
`termination of translation. A start codon would be found upstream of the region
`
`shown above.
`
`31. Depending on where translation begins, each mRNA sequence can be
`
`translated in any one of several “reading frames,” i.e., the various ways in which
`
`the sequence may be divided into sets of nucleotide triplets.
`
`32. Folding: As part of the process of translation, the polypeptide chain
`
`folds to take on its final structure and to become an assembled, active protein.
`
`Folding allows the polypeptide to form its three dimensional structure and occurs
`
`as amino acids within a polypeptide chain interact with one another. Disulfide
`
`bonds or bridges (which are covalent bonds formed between cysteine amino acids)
`
`10
`
`

`
`
`form a scaffolding that helps maintain a protein’s proper three dimensional
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`structure. (Disulfide bonds may also be referred to as S-S bonds.) Such
`
`scaffolding may be present in both simple and more complex proteins, and where
`
`present is essential to the activity of a protein.
`
`33. Post-translational Modification: This generally refers to processes by
`
`which cells may modify a polypeptide after it has been produced to achieve a
`
`mature product, and can include any number of processes.
`
`34. Monomeric and Multimeric Proteins: The most simple proteins are
`
`monomeric, meaning they are formed by only a single polypeptide chain. Other
`
`proteins exist as a complex of multiple polypeptide chains called “multimeric
`
`proteins.” Multimeric proteins can be made up of multiple identical polypeptide
`
`chains or a combination of different polypeptide chains. For multimeric proteins,
`
`amino acid interactions between individual chains and disulfide bonding between
`
`individual chains are critical to correct folding.
`
`35.
`
`Insulin: There are many different types of multimeric proteins formed
`
`from more than one type of polypeptide chain. One example is insulin, which is a
`
`relatively simple and small (i.e., ~ 5,800 Daltons) multimeric protein. (A Dalton is
`
`a standard unit of measurement used to characterize the mass of a protein.) The
`
`insulin molecule contains two different polypeptide chains – an “A” chain
`
`11
`
`

`
`
`consisting of 21 amino acids and a “B” chain consisting of 30 amino acids – that
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`are chemically attached via two inter-chain disulfide (S-S) bonds.
`
`36. The relatively simple configuration of the insulin protein (including its
`
`disulfide bonding scheme) is shown in the illustration below:
`
`(Ex. 2083, Protein Structure, Boundless,
`
`https://www.boundless.com/biology/textbooks/boundless-biology-
`
`textbook/biological-macromolecules-3/proteins-56/protein-structure-304-11437/.
`
`
`
`(last visited Dec. 17, 2016).)
`
`37. As can be seen above, the two chains that comprise insulin are joined
`
`together by two inter-chain disulfide bonds; there is also one intra-chain disulfide
`
`bond.
`
`38. Antibodies: Antibodies, also known as immunoglobulins, are large
`
`tetrameric proteins (which may form even larger complexes) that are expressed and
`
`secreted by B cells (a type of white blood cell made in the bone marrow). There
`
`12
`
`

`
`
`are five classes of antibodies (IgG, IgD, IgE, IgA, and IgM), each of which is
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`further divided into multiple different types called isotypes.
`
`39. A naturally occurring tetrameric antibody is composed of four
`
`polypeptide chains – two identical “heavy” chains (or “H” chains) and two
`
`identical “light” chains (or “L” chains). The heavy and light chains differ in their
`
`size, and thus, their respective molecular weights. By way of example, in
`
`antibodies of the immunoglobulin G (“IgG1”) class, the longer H chains are
`
`naturally comprised of about 450 amino acids and each have a molecular weight of
`
`about 50,000 Daltons, whereas the shorter L chains are naturally comprised of
`
`about 212 amino acids and each have a molecular weight of about 25,000 Daltons.
`
`40. The heavy and light chains of an antibody form what is often depicted
`
`schematically as a Y-shaped molecule via multiple intra- and inter-chain disulfide
`
`bonds. For example, in the case of an antibody of the IgG1 class, there are 12
`
`intra-chain disulfide bonds and 4 inter-chain disulfide bonds that together provide
`
`inter- and intra-chain “scaffolding” within the structure of an antibody, as
`
`discussed above.
`
`41. The below figure (taken from FIG. 1 of the Cabilly ’415 patent)
`
`provides a representation of an antibody’s Y-shape and how the various heavy and
`
`light chains assemble via disulfide bonds to form a functional antibody:
`
`13
`
`

`
`
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`
`42. As shown above, both the heavy and light chains have a “constant”
`
`region and a “variable” region. The “constant” regions may be the same between
`
`different types of antibodies. By contrast, the “variable” regions differ from one
`
`individual antibody to the next; these variable regions are responsible for
`
`identifying and binding to a particular antigen (i.e., a substance that causes the
`
`immune system of an organism to generate antibodies that bind the antigen in order
`
`to provoke an immune response).
`
`43.
`
`In sum, an antibody is a much larger and more complex protein than
`
`insulin (and most other proteins). The molecular weight of a typical antibody is
`
`approximately 150,000 Daltons, or more than 25 times the size of insulin. Further,
`
`the three-dimensional shape of an antibody is more complex and requires much
`
`more post-translational processing.
`
`14
`
`

`
`44. The table below provides a to-scale comparison that illustrates the
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`
`
`relative size of insulin compared to an antibody.
`
`Insulin
`dimer (ab)
`
`
`Immunoglobulin G
`tetramer (a2b2)
`
`
`Structure
`
`
`
`Size
`
`
`
`
`1324 amino acids
`150,000 Daltons
`
`16
`
`
`51 amino acids
`5,800 Daltons
`
`
`
` 3
`
`Disulfide
`Bonds
`
`
`45. At my request, the space-filling models in the above figure were
`
`generated by scientists at Genentech using PyMOL software (available for
`
`download at https://www.pymol.org/). Each sphere represents an atom, with gray
`
`representing carbon, red representing oxygen, blue representing nitrogen, yellow
`
`representing sulfur, and green representing zinc. The size difference between
`
`insulin and an antibody represented above is consistent with my general
`
`15
`
`

`
`
`understanding regarding the relative overall shape and size of each. (See also Ex.
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`2084, RCSB Protein Databank, Molecular Machinery: A Tour of the Protein Data
`
`Bank, http://cdn.rcsb.org/pdb101/learn/resources/2014-mol-mach-poster.pdf (last
`
`visited Dec. 17, 2016) (illustrating the diversity of proteins, with an antibody
`
`shown as protein 13 and insulin as protein 16).)
`
`B. Antibody Production Techniques As Of April 1983
`46. Polyclonal Antibodies: As of April 1983, it was well known that
`
`antibodies could be produced by immunizing an animal with a foreign antigen, and
`
`then recovering the antibodies that had been produced by the animal. Under this
`
`approach, a “polyclonal” serum is generated, i.e., a mixture of antibodies with
`
`varying specificities.
`
`47. As of April 1983, polyclonal antibodies were being widely used to
`
`study the structure and function of antibodies. Due to their varying specificities,
`
`however, polyclonal antibodies had limited usefulness for therapeutic and non-
`
`therapeutic applications. (Ex. 1001 at 1:51-63, 2:40-43.)
`
`48. Hybridomas: In the 1970s, Drs. Georges Köhler and César Milstein
`
`pioneered a technique for producing “monoclonal” antibodies, i.e., antibodies that
`
`have the same amino acid sequence and bind to the same location on the antigen
`
`(called an epitope) in the same way. Their technique involved the use of fused
`
`cells known as “hybridomas.” (See Ex. 2013, G. Kohler & C. Milstein,
`
`16
`
`

`
`
`Continuous Cultures of Fused Cells Secreting Antibody of Predefined Specificity,
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`Nature 256:495-497 (Aug. 7, 1975).)
`
`49. Like the production of polyclonal antibodies, the production of a
`
`hybridoma starts with the immunization of an animal with a particular antigen of
`
`interest. B cells from the animal are then isolated and fused to an immortalized
`
`cell, called a myeloma, which are blood cell-derived cancer cells that have the
`
`ability to grow indefinitely in a cell culture. The fused cells, i.e., hybridomas,
`
`grow continuously in culture and produce the desired antibody.
`
`50.
`
`In the early 1980s, the hybridoma technique was being widely used to
`
`produce monoclonal antibodies, and researchers were focusing on expanding their
`
`use even further. (Ex. 1039, Milstein, Monoclonal Antibodies from Hybrid
`
`Myelomas, Proceedings of the Royal Society of London, 211:393-412, 407 (1981)
`
`(“use of hybrid myelomas to define the complete repertoire of antibodies to single
`
`antigens . . . is now expanding very rapidly” and “many commercial companies are
`
`beginning to market them”); Ex. 2020, Foote Dep. 37, 48 (describing hybridoma
`
`technique as a “very big” deal in the early 1980s due to “significant
`
`achievements”); Ex. 1001 at 1:64-2:11.)
`
`17
`
`
`
`

`
`
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`C. Use of Recombinant Gene Expression To Produce Proteins
`1.
`Basic principles of recombinant gene expression
`51. Recombinant gene expression is a method that allows for the
`
`production and isolation of a protein of interest in a foreign, i.e., “heterologous”
`
`host organism, usually a cell or “host cell.”
`
`52. As explained in the Cabilly ’415 patent, this process involves four
`
`fundamental steps: (1) identification and isolation of a particular gene of interest
`
`(Ex. 1001 at 4:10-12), (2) insertion of that DNA sequence into a vector or plasmid
`
`(id. at 4:17-21), (3) insertion of that vector or plasmid into a suitable host cell (id.
`
`at 4:20), and (4) expression of the sequence, i.e., transcribing the gene of interest
`
`into mRNA, and then translating the mRNA into a polypeptide, by the host cell.
`
`(Id. at 4:23-29.) Host cells transformed with the vector are grown in culture to
`
`express the gene of interest in high quantities and the resulting protein is then
`
`isolated. (Id. at 4:21-24.)
`
`53. Vectors: A vector, comprised of DNA, includes a number of
`
`components to assist with the insertion and expression of the DNA sequence of
`
`interest. For example, a vector will generally include a number of “restriction
`
`enzyme sites,” which are locations in DNA that can be cut by certain proteins
`
`known as restriction enzymes. Restriction enzyme sites allow for the insertion of
`
`the DNA sequence of interest into the vector. (E.g., Ex. 2085, Vectors: A survey
`
`18
`
`

`
`
`of molecular cloning vectors and their uses, Chapter 1: The Plasmid pBR322,
`
`Case No. IPR2016-00710
`Declaration of John Fiddes, Ph.D.
`
`Rodriguez, R.L. and Denhardt, D.T. eds. 1988).)
`
`54. Promoters/Terminators: A vector also includes regulatory sequences
`
`such as promoters and terminators to signal the initiation and termination of
`
`transcription. (Ex. 1001 at 8:57-9:15, 9:56-10:3.) A promoter is a specific DNA
`
`sequence that signals where RNA polymerase should bind the template DNA and
`
`where mRNA synthesis should begin. A transcription terminator is a specific
`
`DNA sequence that signals where mRNA synthesis should end.
`
`55. Markers: A vector or plasmid will also often include a “marker” (also
`
`called a “marker gene”) that allows for

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