throbber
Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`
`Filed on behalf of Patent Owner Genentech, Inc. by:
`
`David L. Cavanaugh (Reg. No. 36,476)
`Rebecca A. Whitfield (Reg. No. 73,756)
`Robert J. Gunther, Jr. (Pro Hac Vice)
`Lisa J. Pirozzolo (Pro Hac Vice)
`Kevin S. Prussia (Pro Hac Vice)
`Andrew J. Danford (Pro Hac Vice)
`WILMER CUTLER PICKERING
` HALE AND DORR LLP
`1875 Pennsylvania Ave., NW
`Washington, DC 20006
`
`
`Adam R. Brausa (Reg. No.
`60,287)
`Daralyn J. Durie (Pro Hac
`Vice)
`DURIE TANGRI LLP
`217 Leidesdorff Street
`San Francisco, CA 94111
`
`
`UNITED STATES PATENT AND TRADEMARK OFFICE
`
`____________________________________________
`
`BEFORE THE PATENT TRIAL AND APPEAL BOARD
`
`____________________________________________
`
`CELLTRION, INC.,
`Petitioner,
`
`v.
`
`GENENTECH, INC.,
`Patent Owner.
`____________________________________________
`
`Case IPR2017-01122
`Patent Nos. 7,892,549
`____________________________________________
`
`EXPERT DECLARATION OF ROBERT S. KERBEL, PH.D.
`
`
`
`Genentech 2061
`Celltrion v. Genentech
`IPR2017-01122
`
`

`

`
`
`
`
`I.
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`TABLE OF CONTENTS
`
`Page
`
`INTRODUCTION AND BACKGROUND .................................................. 1
`
`A. Qualifications and Experience ............................................................ 3
`
`B.
`
`C.
`
`Compensation ................................................................................... 11
`
`Prior Expert Testimony ..................................................................... 11
`
`II.
`
`STATE OF THE ART OF PRECLINICAL CANCER RESEARCH AS OF
`1997 ............................................................................................................ 12
`
`A.
`
`In Vitro Testing ................................................................................ 12
`
`B.
`
`In Vivo Testing ................................................................................. 13
`
`1.
`
`2.
`
`Early Mouse Models ............................................................... 13
`
`Development of Human Xenografts ........................................ 15
`
`C.
`
`Xenograft Models in the 1990s ......................................................... 17
`
`III. THE PRECLINICAL DATA RELIED UPON BY PETITIONER AND DR.
`EARHART ................................................................................................. 20
`
`IV. SUMMARY AND BASES FOR OPINION ............................................... 22
`
`A.
`
`B.
`
`C.
`
`It was well known in the mid to late 1990s, and remains true
`today, that preclinical results are not a reliable predictor of
`human clinical results. ...................................................................... 24
`
`It was well-known that a single tumor line, as used in the
`Baselga references, could provide skewed results. ............................ 27
`
`It was well-known that preclinical data does not predict toxicity
`in human patients due to tissue differences. ...................................... 32
`
`i
`
`

`

`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`The preclinical studies in the Baselga references were further
`limited by the manner of creating the xenograft models. ................... 35
`
`The end point in the Baselga Abstracts of measuring tumor
`response rate does not provide information on time to disease
`progression. ...................................................................................... 37
`
`D.
`
`E.
`
`
`
`
`
`ii
`
`

`

`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`
`I.
`
`INTRODUCTION AND BACKGROUND
`
`1.
`
`I, Robert S. Kerbel, Ph.D., have been retained by counsel for
`
`Genentech, Inc. (“Patent Owner”) as an expert in Celltrion, Inc. v. Genentech, Inc.,
`
`IPR2017-01122, challenging claims 1-11 and 14-18 of U.S. Patent No. 7,892,549
`
`(the “’549 patent”). I understand that on October 4, 2017, the Patent Trial and
`
`Appeal Board (the “Board”) instituted inter partes review as to these claims of the
`
`’549 patent.
`
`2. As described in more detail below, I understand that the ’549 patent is
`
`directed to treatment of a certain type of breast cancer that overexpresses a human
`
`epidermal growth factor, Erb-B2 receptor tyrosine kinase (also called HER2). I
`
`further understand that the ’549 patent describes a treatment therapy that includes
`
`at least an anti-erbB2 antibody (specifically, rhuMAb HER2 and now known as
`
`Herceptin® or trastuzumab), a taxoid (specifically, paclitaxel), and a third agent in
`
`an amount effective to extend the time to disease progression in a human patient.
`
`3.
`
`I further understand that the Petitioner Celltrion, Inc. (“Petitioner”)
`
`makes certain arguments, and that its expert Dr. Robert Howard Earhart, Jr. asserts
`
`certain opinions, regarding the predictive value of preclinical studies in obtaining
`
`specific results in clinical studies. In particular, those preclinical studies are
`
`described in two one-paragraph abstracts, J. Baselga et al., Anti HER2 Humanized
`
`Monoclonal Antibody (MAb) Alone and in Combination with Chemotherapy
`
`1
`
`

`

`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`
`Against Human Breast Carcinoma Xenografts, 13 PROC. AM. SOC. CLINICAL
`
`ONCOLOGY 63 (Abstract 53) (1994) (Ex. 1019), and Baselga et al., Antitumor
`
`Activity of Paclitaxel in Combination with Anti-growth Factor Receptor
`
`Monoclonal Antibodies in Breast Cancer Xenografts, 35 PROC. AM. ASSOC.
`
`CLINICAL CANCER RESEARCH 380 (Abstract 2262) (Ex. 1021) (collectively, the
`
`“Baselga Abstracts”). These preclinical studies are also mentioned in J. Baselga et
`
`al., Phase II Study of Weekly Intravenous Recombinant Humanized Anti-p185HER2
`
`Monoclonal Antibody in Patients with HER2/neu-Overexpressing Metastatic
`
`Breast Cancer, 14(3) J. CLINICAL ONCOLOGY 737 (1996) (“Baselga ʼ96”) (Ex.
`
`1020).
`
`4.
`
`I have been asked to review these opinions and publications and to
`
`offer my own opinion as to whether, in the 1997 time frame, the preclinical data
`
`described in these references reporting on testing a combination of rhuMAb HER2
`
`and paclitaxel in a xenograft mouse model suggests that the same combination in
`
`humans would extend the time to disease progression without an increase in severe
`
`adverse effects. As explained in detail below, the preclinical study described in
`
`these references would not have supported any testing or suggested successful
`
`results in human patients given the known limitations of mouse models at that time
`
`and in light of the design of the experiments at issue in particular.
`
`2
`
`

`

`
`
`5. A list of materials I have reviewed in preparation of this Declaration is
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`attached as Exhibit B. I have also relied upon my scientific knowledge as of
`
`December 1997, which I have been told is the relevant time period for viewing the
`
`state of the art.
`
`A. Qualifications and Experience
`
`6. My background is summarized below and in my curriculum vitae,
`
`which includes a list of my publications and is attached as Exhibit A.
`
`7.
`
`I am currently a Senior Scientist in the Biological Sciences Platform of
`
`the Sunnybrook Research Institute, which is affiliated with the Sunnybrook Health
`
`Sciences Centre, a University of Toronto-affiliated teaching hospital. I also hold a
`
`cross appointment as a full Professor in the Department of Medical Biophysics at
`
`the University of Toronto.
`
`8.
`
`I earned a Bachelor of Sciences (BSc) degree in general sciences in
`
`1967 at the University of Toronto. I received a Ph.D. in immunology from the
`
`Department of Microbiology and Immunology, Queen’s University in Kingston,
`
`Ontario in 1972. I then undertook a two-year period of postdoctoral training at the
`
`Chester Beatty Research Institute, Institute for Cancer Research in London
`
`England where I continued my studies in basic immunology. It was also here
`
`where I first became interested in tumor biology and tumor immunotherapy, and
`
`hence preclinical experimental therapeutics.
`
`3
`
`

`

`
`
`9. After completing my postdoctoral training in 1975, I took a position as
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`an Assistant Professor in the Department of Pathology, Queen’s University in
`
`Kingston, Ontario, as a member of a dedicated cancer research group, supported
`
`primarily by the National Cancer Institute of Canada. I was also a recipient of a
`
`career award called Research Scholar of the National Cancer Institute of Canada
`
`which covered my salary. I remained there for the next decade, during which time
`
`I became interested in immunotherapy for treatment of cancer, including for
`
`metastatic disease, and in developing improved preclinical mouse cancer models
`
`for this purpose. I was appointed Director of the Cancer Research Group in 1981.
`
`At this time I became a Research Associate of the National Cancer Institute of
`
`Canada, a more senior career award.
`
`10.
`
`In 1985, I moved to Toronto to take up a position as Director of the
`
`Cancer and Cell Biology division in a new research institute at Mt. Sinai Hospital,
`
`a University of Toronto teaching hospital. I continued to serve in this capacity
`
`until 1991 at which time I moved to my present location at the Sunnybrook Health
`
`Sciences Centre, where I was recruited as Director of a cancer research division
`
`that was one of four major research components of a new research institute
`
`currently called the Sunnybrook Research Institute (SRI). I served in this
`
`leadership capacity until 2001. During this period, the Cancer Research Division
`
`was incorporated into a broader program called Biological Sciences. The cancer
`
`4
`
`

`

`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`
`research component of this program was affiliated with the Toronto-Sunnybrook
`
`Regional Cancer Centre, one of the largest cancer treatment centres in Canada,
`
`which was (and still is) located adjacent to my laboratory, and with which I had a
`
`close affiliation.
`
`11. During my career, my major research interests have included studies in
`
`tumor immunology, metastasis, tumor angiogenesis, antiangiogenic therapy,
`
`chemotherapy, drug resistance, and molecular targeted therapies.
`
`12. Especially relevant to these proceedings, my research has included
`
`preclinical studies involving antibodies to the EGF receptor (C225/cetuximab or
`
`Erbitux®) as well as to HER2, using the anti-erbB2 antibody known as
`
`4D5/trastuzumab (Herceptin®), Ras inhibitors, and proteosome inhibitors. For
`
`example, from the 1990s through the present, my research has included learning
`
`the properties and mechanisms of action of rhuMAb HER2 (see, e.g., Ex. 2077,
`
`Alicia M. Viloria Petit, et al., Neutralizing Antibodies against Epidermal Growth
`
`Factor and ErbB-2/neu Receptor Tyrosine Kinases Down-Regulate Vascular
`
`Endothelial Growth Factor Production by Tumor Cells in Vitro and in Vivo,
`
`151(6) AM. J. OF PATHOLOGY 1523 (1997); Ex. 2078, Jeanne M. du Manoir, et al.,
`
`Strategies for Delaying or Treating In vivo Acquired Resistance to Trastuzumab in
`
`Human Breast Cancer Xenografts, 12(3) CLINICAL CANCER RESEARCH 904
`
`(2006)), as well as exploring therapies involving trastuzumab with chemotherapy.
`
`5
`
`

`

`
`(See, e.g., Ex. 2079, Giulio Francia, et al., Long-Term Progression and
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`Therapeutic Response of Visceral Metastatic Disease Non-Invasively Monitored in
`
`Mouse Urine Using β-Human Choriogonadotropin Secreting Tumor Cell Lines,
`
`7(10) MOLECULAR CANCER THERAPY 3452 (2008); Ex. 2080, Giulio Francia, et al.,
`
`Comparative Impact of Trastuzumab and Cyclophosphamide on HER-2–Positive
`
`Human Breast Cancer Xenografts, 15(20) CLINICAL CANCER RESEARCH 6358
`
`(2009).)
`
`13.
`
`I have also worked with a number of different chemotherapy drugs
`
`over the years such as cyclophosphamide, cisplatin, vinblastine, topotecan, as well
`
`as paclitaxel and a nanomedicine formulation of paclitaxel called Nab-paclitaxel.
`
`For example, over the last twenty years, I have conducted research—and published
`
`a number of papers—involving the use of paclitaxel in human tumor xenografts of
`
`breast or ovarian cancer. (See, e.g., Ex. 2081, Eric Guerin, et al., A Model of
`
`Postsurgical Advanced Metastatic Breast Cancer More Accurately Replicates the
`
`Clinical Efficacy of Antiangiogenic Drugs, 73(9) CANCER RESEARCH 2743 (2013);
`
`Ex. 2082, Sylvia S.W. Ng, et al., Influence of Formulation Vehicle on Metronomic
`
`Taxane Chemotherapy: Albumin-Bound versus Cremophor EL-Based Paclitaxel
`
`12(14) CLINICAL CANCER RESEARCH 4331 (2006); Ex. 2083, Giannoula Klement,
`
`et al., Differences in Therapeutic Indexes of Combination Metronomic
`
`6
`
`

`

`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`
`Chemotherapy and an Anti-VEGFR-2 Antibody in Multidrug-resistant Human
`
`Breast Cancer Xenografts, 8 CLINICAL CANCER RESEARCH 221 (2002).)
`
`14.
`
` All of the preclinical work I have done in the area of experimental
`
`therapeutics has involved assessment of response and resistance to most of the
`
`various types of aforementioned drugs, utilizing a variety of mouse models to
`
`assess the therapeutic impact on metastatic disease as well as on established
`
`primary tumors. Many of these models involve “orthotopic” transplantation of
`
`cells from established human tumor cell lines (i.e., transplantation of tumor cells
`
`into the organ from which the cancer under study was derived such as breast
`
`cancer cells injected into the mouse mammary fat pads), to create orthotopic
`
`human tumor xenografts.
`
`15. Some examples of the work I did and published in the early to mid-
`
`1990s on orthotopic transplantation of human tumor cell lines include injection of
`
`human malignant melanoma cells subdermally rather than subcutaneously (see,
`
`e.g., Ex. 2085, Maria Rosa Bani, et al., Multiple Features of Advanced Melanoma
`
`Recapitulated in Tumorigenic Variants of Early Stage (Radial Growth Phase)
`
`Human Melanoma Cell Lines: Evidence for a Dominant Phenotype, 56 CANCER
`
`RESEARCH 3075 (1996); Ex. 2086, Hiroaki Kobayashi, et al., Variant Sublines of
`
`Early-Stage Human Melanomas Selected for Tumorigenicity in Nude Mice Express
`
`a Multicytokine-Resistant Phenotype, 144(4) AM. J. OF PATHOLOGY 776 (1994)),
`
`7
`
`

`

`
`and human bladder cancer cells into the bladder. (See, e.g., Ex. 2087, Robert S.
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`Kerbel, et al., Importance of Orthotopic Transplantation Procedures in Assessing
`
`the Effects of Transfected Genes on Human Tumor Growth and Metastasis, 10
`
`CANCER AND METASTASIS REVIEWS 201 (1991); Ex. 2088, Theodorescu, D., et al.,
`
`Lack of Influence of c-Ha-ras Expression in the Drug Sensitivity of Human
`
`Bladder Cancer Histocultured in Three-Dimensions, 13 ANTICANCER RESEARCH
`
`941 (1993).)
`
`16. Throughout my career and in the course of this research, I have
`
`worked with medical oncologists in two broad complimentary ways. First, I have
`
`had medical (and surgical) oncologists train in my laboratory as Ph.D. students or
`
`postdoctoral fellows. Second, I have collaborated with many medical oncologists
`
`on clinical trials as well as on preclinical research in some cases to support those
`
`trials. Some examples of the former include clinical trials involving an
`
`investigational concept known as low-dose ‘metronomic’ chemotherapy, which
`
`resulted in co-authored publications (see, e.g., Ex. 2089, Nan Soon Wong, et al.,
`
`Phase I/II trial of Metronomic Chemotherapy with Daily Dalteparin and
`
`Cyclophosphamide, Twice-Weekly Methotrexate and Daily Prednisone as Therapy
`
`for Metastatic Breast Cancer Using Vascular Endothelial Growth Factor and
`
`Soluble Vascular Endothelial Growth Factor Receptor Levels as Markers of
`
`Response, 28 J. CLINICAL ONCOLOGY 723 (2010); Ex. 2090, Rena Buckstein, et al.,
`
`8
`
`

`

`
`Lenalidomide and Metronomic Melphalan for CMML and Higher Risk MDS: A
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`Phase 2 Clinical Study with Biomarkers of Angiogenesis, 38 LEUKEMIA RESEARCH
`
`756 (2014)), and the antiangiogenic drug bevacizumab (Avastin®). (See, e.g., Ex.
`
`2091, Julia L. Glade Bender, et al., Phase I Trial and Pharmacokinetic Study of
`
`Bevacizumab in Pediatric Patients with Refractory Solid Tumors: A Children’s
`
`Oncology Group Study, 26(3) J. CLINICAL ONCOLOGY 399 (2008).)
`
`17. Also, in my capacity as director of the cancer research division at
`
`Sunnybrook from 1991-2001, I had the opportunity to interact with medical,
`
`surgical and radiation oncologists in the Toronto-Sunnybrook Regional Cancer
`
`Centre located at Sunnybrook (and now called the Odette Cancer Centre). One of
`
`my responsibilities was to foster interactions between basic cancer and
`
`translational researchers such as myself and the clinical oncologists. To this end, I
`
`organized occasional evening research-in-progress meetings between the two
`
`groups. One of the collaborators was Dr. Joyce Slingerland, a breast cancer
`
`medical oncologist, whom I recruited into the cancer research division when I was
`
`Director, and who held a cross appointment at the Toronto-Sunnybrook Regional
`
`Cancer Centre.
`
`18. Since 1971, I have authored or co-authored 423 published papers. A
`
`number of these papers were peer reviewed invited reviews requested by the
`
`editors of journals such as Science, Nature, the New England Journal of Medicine,
`
`9
`
`

`

`
`Cancer Cell, and a number of review journals such as Nature Reviews Clinical
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`Oncology, Nature Reviews Cancer, and Nature Reviews Drug Discovery. Many of
`
`these reviews or editorial commentaries dealt with experimental therapeutics,
`
`including commentaries on preclinical mouse tumor models for assessing cancer
`
`drugs. I also have been invited, over my career, to give almost 900 lectures around
`
`the world at scientific meetings, universities, research institutes, clinical cancer
`
`treatment centres, courses, and advisory boards of companies during my career.
`
`Many of these were Keynote or Plenary lectures, as noted in my c.v.
`
`19.
`
`I have been active in advisory activities, both in industry and
`
`academia. My numerous industry associated advisory roles as a member of
`
`scientific advisory boards (SABs) or as a consultant have dealt mainly with
`
`experimental therapeutics, and preclinical models to assess cancer drug activity in
`
`mice.
`
`20. Over the course of my career I have received competitive grant support
`
`from numerous agencies including the National Institute of Health, USA which I
`
`held as a foreign investigator for 27 continuous years, the Canadian Institute for
`
`Health Research (over 40 years, continuously), the Canadian Cancer Society
`
`Research Institute (35 years continuously), the Canadian Breast Cancer Foundation
`
`(6 years of support), the Ontario Institute for Cancer Research, and Worldwide
`
`Cancer Research.
`
`10
`
`

`

`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`
`
`21. Awards I have received include The Robert L. Noble Prize for
`
`Excellence in Cancer Research by the Canadian Cancer Society and National
`
`Cancer Institute of Canada in 2004, the Breast Cancer Research Award from the
`
`European Institute of Oncology in 2008, a Man of Distinction award from the
`
`Israel Cancer Research Fund in 2011, and the Colin Thomson Memorial Medal for
`
`achievements in cancer research from Worldwide Cancer Research (formerly
`
`known as the Association for International Cancer Research) in 2013. Throughout
`
`my career I have also received a succession of career awards (two of which I have
`
`already noted) including a Terry Fox Career Scientist award from the National
`
`Cancer Institute of Canada (1986-1997), a Canada Research Chair in Tumor
`
`Biology, Angiogenesis, and Antiangiogenic Therapy from the Canadian
`
`government (2001-2015), and an endowed professorship, the John & Elizabeth
`
`Tory Family Chair in Experimental Oncology (1993-2001).
`
`B. Compensation
`
`22.
`
`I am being compensated at my normal consulting rate for my work,
`
`which is $600 per hour. My compensation is not dependent on and in no way
`
`affects the substance of my statements in this Declaration.
`
`C.
`
`23.
`
`Prior Expert Testimony
`
`I have not provided expert testimony within the last four years.
`
`Concurrently with this declaration, I am submitting declarations in Hospira, Inc. v.
`
`11
`
`

`

`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`
`Genentech, Inc., IPR2017-00731, Hospira, Inc. v. Genentech, Inc., IPR2017-
`
`00737, and Celltrion, Inc. v. Genentech, Inc., IPR2017-01121.
`
`II.
`
`STATE OF THE ART OF PRECLINICAL CANCER RESEARCH AS
`OF 1997
`
`24. Preclinical cancer research provides the foundation for subsequent
`
`clinical trials by identifying drugs of interest that appear to act on human cancer
`
`cells. Generally, preclinical research provides information on (a) whether and how
`
`a particular drug acts on tumor cells to slow their growth and/or shrink the tumors,
`
`(2) the mechanism of action of the drug (i.e., how the drug interacts with cancer
`
`cells or other cells in the tumor microenvironment such as blood vessel or immune
`
`cells to slow growth and/or kill cells), and (3) whether and how the drug affects
`
`non-cancerous cells, especially with respect to causing host toxicities. Based on
`
`the results of preclinical research, academic clinicians, basic scientists, and
`
`clinicians working for drug companies decide whether to propose testing the drug
`
`in humans. This testing of a drug in humans is called clinical trials.
`
`25. There are two main or basic types of preclinical research models: in
`
`vitro (cell cultures), and in vivo (animal subjects). Typically, both in vitro and in
`
`vivo studies are performed before progressing to human clinical trials.
`
`A.
`
`26.
`
`In Vitro Testing
`
` In in vitro testing, established “cell lines” are often used, meaning that
`
`all of the cells derive from a single cancer. These established (also called
`
`12
`
`

`

`
`“permanent” or “continuous”) cell lines can be from a human patient and
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`sometimes are also genetically modified in the laboratory. Even when the cell
`
`lines are derived from a patient, the cells can differ substantially from cancer cells
`
`in humans as a result of the cell lines adapting to being cultured on plastic and
`
`being maintained for long periods in vitro. And although the individual cells in a
`
`cell line may have some variations due to genetic mutations or epigenetic changes,
`
`these variations differ in degree and content compared to cancer cells in human
`
`patients.
`
`27. Despite the differences between established cell lines and cancer cells
`
`in patients, in vitro testing was in 1997 and is still today useful because researchers
`
`can test, with relative efficiency, multiple different cell lines representing different
`
`types of cancers (e.g., colon cancer, lung cancer, etc.), as well as different lines of
`
`the same general type of cancer (e.g., different cell lines of HER2-positive breast
`
`cancer). In cancer research, common end points for testing potential cancer drugs
`
`include relative inhibition or proliferation of the cancer cells (i.e., how much does
`
`the drug prevent the cancer cells from spreading?), and cell death (i.e., does the
`
`drug kill the cancer cells?).
`
`B.
`
`In Vivo Testing
`
`1.
`
`Early Mouse Models
`
`13
`
`

`

`
`
`28.
`
` In contrast to in vitro testing, in vivo testing involves using animal
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`models, by far the most common of which is the tumor-bearing mouse.
`
`29. Up until about 1980, these models mostly involved the transplantation
`
`of mouse tumor cells or fragments of mouse tumor tissue into strains of
`
`immunocompetent mice that are genetically compatible with the donor tumor cell
`
`population or tissue.
`
`30. The tumor cells injected were almost always obtained from established
`
`cell lines grown in tissue culture (i.e., in vitro), and were most often injected
`
`subcutaneously in the hind leg or flank of the mouse. This is called ectopic
`
`transplantation for most types of tumors, i.e., those that do not normally arise in the
`
`skin – which is virtually all types of cancer. By injecting tumor cells into the
`
`flank, the resultant “primary” tumors would grow quickly and could be
`
`subsequently measured by using vernier calipers.
`
`31. Sometimes the cells were injected intravenously to generate
`
`experimental metastases, mainly in the lungs. Measuring the cancer metastases,
`
`however, required sacrificing the mouse to visually inspect the lungs.
`
`32.
`
` Sometimes the tumor cells were injected “orthotopically”, i.e., into the
`
`organ from which the cancer cell population was derived, e.g., breast cancer cells
`
`are injected into mammary fat pads of female mice, to generate “primary” tumors.
`
`14
`
`

`

`
`
`33. The impact of a cancer therapy on tumor growth in the primary tumor
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`models was assessed by measuring changes in tumor volume/size, and sometimes
`
`changes in survival times via comparison to various control groups.
`
`2.
`
`Development of Human Xenografts
`
`34. Starting in earnest in the 1970s, researchers started to develop and
`
`utilize human tumor xenografts as animal models to test cancer drugs. This was
`
`mainly made possible by the discovery of the “nude” athymic mouse which cannot
`
`develop thymus-derived T lymphocytes – the main cell type responsible for
`
`rejecting foreign tissue such as a human skin graft, or in this case injected human
`
`tumor cell or tissue graft. Prior to that it was possible to create T cell deficient
`
`mice using a combination of thymectomy plus radiation or chemotherapy, but they
`
`were not used widely for human tumor xenograft studies.
`
`35.
`
`In a xenograft, dispersed human cancer cells or intact human tumor
`
`tissue fragments are transplanted into immunocompromised mice (such as athymic
`
`nude mice). The rationale behind using such xenografts is that they more closely
`
`resemble the cancer in a human cancer patient than tumor cells from established
`
`cell cultures or mouse tumors, and therefore the reactions and results caused by
`
`drugs or therapies using such human tumor xenografts approximate somewhat
`
`better what may occur in humans.
`
`15
`
`

`

`
`
`36.
`
`Just as with the early mouse tumor models, with a human tumor
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`xenograft model the cancer cells injected into the mouse will form tumors, which
`
`researchers can measure and monitor when treating the mice with a drug. The
`
`most common endpoint for testing drugs in these xenograft models is tumor
`
`response (also called response rate), which measures whether the tumors continue
`
`to progress normally or respond in some other way to treatment, whether by being
`
`delayed in growth, shrinking in size, or becoming fully eradicated. In the 1990s,
`
`and beforehand, xenograft model results rarely used or reported progression free
`
`survival (PFS) or time to progression (TTP) as endpoints.
`
`37. When xenograft models were first developed and through the mid-
`
`1980s, there was excitement about the possibility that they could be used to help
`
`select a particular drug therapy for an individual patient by using a sample of the
`
`cancer taken directly from the human patient to create an ‘individualized’
`
`xenograft. This individualized xenograft model, it was theorized, could be highly
`
`accurate in predicting cancer drug activity in individual respective patients, and
`
`thus it was hypothesized that this model could be used as a “predictive biomarker”
`
`for selecting drug therapies tailored to the patient who donated the tumor sample.
`
`Indeed, a number of studies reported strong correlations between the tumor
`
`response in mice as compared to the human patient from which the tumor
`
`specimen was obtained. (Ex. 1026, H.H. Fiebig, et al., Comparison of Tumor
`
`16
`
`

`

`
`Response in Nude Mice and in the Patients, 74 BEHRING INST. MITT. 343, 343,
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`349-350 (1984) (hereafter “Fiebig 1984”); Ex. 2092, Beppino C. Giovanella, et al.,
`
`Correlation Between Response to Chemotherapy of Human Tumors in Patients and
`
`in Nude Mice, 52 CANCER 1146, 1146 (1983).)
`
`38. Yet it turned out that these “patient derived” tissue xenografts were not
`
`predictive of treatment for other human patients having the same type of cancer.
`
`Further, these patient-derived xenografts had many serious technical problems,
`
`including the low percentages of tumor specimens for most tumor types that “took”
`
`and grew progressively in the immune deficient mice, and the generally long
`
`latency periods for some tumors to grow and become visible (e.g., 4-6 months)
`
`before treatment could be started and the impact assessed. (Ex. 1026, Fiebig 1984,
`
`346-347.) As a result of these problems, the human patients from which samples
`
`were taken had often died before any of the results of the tests on the xenograft
`
`models could be applied to the patients.
`
`C. Xenograft Models in the 1990s
`
`39. As a result of the problems with a patient-derived xenograft strategy,
`
`researchers focused on the more practical method of using established tumor cell
`
`lines from tissue culture for human tumor xenografts. These tumor cell lines often
`
`had a take rate of 100% (meaning that tumors grew 100% of the time), and tumors
`
`usually appeared within weeks of tumor cell transplantation. The reason for the
`
`17
`
`

`

`
`success in establishing tumors using cell lines instead of samples from human
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`patients is that the cell lines contain a comparatively high fraction of dividing cells
`
`by virtue of being held in culture – those cells that do not divide as quickly are out-
`
`paced by those that do, and this out-pacing is compounded over time in cell
`
`culture.
`
`40. That these cell cultures include more rapidly dividing cells also means,
`
`however, that they do not accurately reflect cancer as found in human patients.
`
`Further, the rapid growth rate of cell cultures (and the resulting xenografts made
`
`from them) means that they have greater sensitivity to virtually all chemotherapy
`
`drugs, which are designed to preferentially target rapidly dividing cells. As a
`
`preclinical model, however, the xenograft nonetheless provides information on
`
`whether and how a cancer line will react to a particular drug, even if it is not
`
`predictive of the particular reaction in a population of human patients with
`
`heterogenous tumors of the same or different types.
`
`41. As with the early mouse models, a xenograft can be formed either
`
`through ectopic or orthotopic transplantation. For example, in the 1990s,
`
`orthotopic xenograft models for breast cancer could be created by injecting breast
`
`cancer cells into the mammary fat pad. (Ex. 2093, L. Bao, et al., Effects of
`
`Inoculation Site and Matrigel on Growth and Metastasis of Human Breast Cancer
`
`Cells, 70 BRITISH J. OF CANCER 228, 229 (1994).) Again, the rationale was that
`
`18
`
`

`

`
`such a model would more closely approximate how the cancer and therapy may act
`
`Case No. IPR2017-01122
`Declaration of Robert S. Kerbel, Ph.D.
`
`in humans.
`
`42. Similar to the established cell lines for in vitro cancer research, there
`
`were multiple cell lines available for human tumor xenograft models, allowing
`
`researchers to test different types of cancers, as well as different lines of the same
`
`type of cancer. For example, HER2-positive breast cancer refers to breast cancer
`
`in which the cells have many more than the usual two copies of the gene encoding
`
`for the epidermal growth factor receptor called ErbB2 (HER2), resulting in
`
`elevated expression of the HER2 epidermal growth factor protein. In the 1990s,
`
`there were several different cell lines of HER2-positive breast cancer, each with
`
`different numbers of the ErbB2 gene in the cells. For example, one study in 1995
`
`(Ex. 2064, János Szöllösi, et al., ERBB-2 (HER2/neu) Gene Copy Number,
`
`p185HER-2 Overexpression, and Intratumor Heterogeneity in Human Breast Cancer,
`
`55 CANCER RESEARCH 5400, 5402 (1995) (hereafter “Szöllösi 1995”)) estimated
`
`that a specific cell line identified as BT-474 ha

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