throbber
PHIGENIX
`PHIGENIX
`Exhibit 1004
`Exhibit 1004
`
`

`
`Cancer Research
`
`
`.m< :;
`
`Targeting HER2-Positive Breast Cancer with
`Trastuzumab-DM1, an Antibody—Cytotoxic Drug Conjugate
`
`Gail D. Lewis Phillips, Guangmin Li, Debra L. Dugger, et al.
`
`Cancer Res 2008;68:9280—9290.
`
`Updated version
`
`Access the most recent version of this article at:
`http://cancerres.aacrjournals.org/content/68/22/9280
`
`Cited Articles
`
`This article cites by 45 articles, 24 of which you can access for free at:
`http://cancerres.aacrjournals.org/content/68/22/9280.full.html#ref—list—1
`
`Citing articles
`
`This article has been cited by 48 HighWire—hosted articles. Access the articles at:
`http://cancerres.aacrjournals.org/content/68/22/9280.full.html#related—urls
`
`E-mail alerts
`
`Sign up to receive free email—alerts related to this article orjournal.
`
`Reprints and
`subscriptions
`
`To order reprints of this article or to subscribe to the journal, contact the AACR Publications
`Department at pubs@aacr.org.
`
`Permissions
`
`To request permission to re—use all or part of this article, contact the AACR Publications
`Department at permissions@aacr.org.
`
`Downloaded from cancerres.aacrjourna|s.org on February 9, 2014. © 2008 American Association for Cancer
`Research.
`
`PH IGEN IX
`
`Exhibit 1004-01
`
`

`
`
`
`Targeting HER2-Positive Breast Cancer with Trastuzumab-DM1,
`
`an Antibody—Cytotoxic Drug Conjugate
`
`Gail D. Lewis Phillips,1 Guangmin Li,1 Debra L. Dugger,1 Lisa M. Crocker,1 Kathryn L. Parsons,1
`Elaine Mai,1 Walter A. Blattler,2 John M. Lambert,2 Ravi VJ. Chari,2 Robert J. Lutz,2
`Wai Lee T. Wong,1 Frederic S. Jacobson,1 Hartmut Koeppen,1 Ralph H. Schwall,1
`Sara R. Kenkare—Mitra,1 Susan D. Spencer,1 and Mark X. Sliwkowskil
`‘Genentech, Inc., South San Francisco, California and ‘ImmunoGen, Inc., Waltham, Massachusetts
`
`Abstract
`
`HER2 is a validated target in breast cancer therapy. Two drugs
`are currently approved for HER2-positive breast cancer:
`trastuzumab (Herceptin), introduced in 1998, and lapatinib
`(Tykerb),
`in 2007. Despite these advances, some patients
`progress through therapy and succumb to their disease. A
`variation on antibody-targeted therapy is utilization of anti-
`bodies to deliver cytotoxic agents specifically to antigen-
`expressing tumors. We determined in vitro and in vivo efficacy,
`pharmacokinetics, and toxicity of trastuzumab-maytansinoid
`(microtubule-depolymerizing agents) conjugates using disul-
`fide and thioether linkers. Antiproliferative effects of trastu-
`zumab-maytansinoid conjugates were evaluated on cultured
`normal and tumor cells. In vivo activity was determined in
`mouse breast cancer models, and toxicity was assessed in rats
`as measured by body weight loss. Surprisingly, trastuzumab
`linked to DMI
`through a nonreducible thioether linkage
`(SMCC), displayed superior activity compared with unconju-
`gated trastuzumab or trastuzumab linked to other maytansi-
`noids through disulfide linkers. Serum concentrations of
`trastuzumab-MCC-DMI remained elevated compared with
`other conjugates, and toxicity in rats was negligible compared
`with free DMI or trastuzumab linked to DMI through a
`reducible linker. Potent activity was observed on all HER2-
`overexpressing tumor cells, whereas nontransformed cells
`and tumor cell
`lines with normal HER2 expression were
`unaffected. In addition, trastuzumab-DMI was active on HER2-
`overexpressing, trastuzumab-refractory tumors. In summary,
`trastuzumab-DMI shows greater activity compared with
`nonconjugated trastuzumab while maintaining selectivity for
`HER2-overexpressing tumor cells. Because trastuzumab linked
`to DMI through a nonreducible linker offers improved efficacy
`and pharmacokinetics and reduced toxicity over the reducible
`disulfide linkers evaluated, trastuzumab-MCC-DMI was select-
`ed for clinical development. [Cancer Res 2008;68(22):9280—90]
`
`Introduction
`
`The HER2 (ErbB2) receptor tyrosine kinase is a member of the
`epidermal growth factor receptor
`family of transmembrane
`
`Note: Current address for H. Koeppen: M. D. Anderson Cancer Center, Houston,
`TX 77030.
`Requests for reprints: Gail D. Lewis Phillips, Genentech, Inc., 1 DNA Way, South
`San Francisco, CA 94080. Phone: 650-225-2201; Fax: 650-225-1411; E-mail: gdl@
`gene.com or Mark K Sliwkowski, Genentech, Inc., 1 DNA Way, South San Francisco,
`CA 94080. Phone: 650-225-1247; Fax: 650-225-5770; E-mail: ma.rks@gene.com.
`©2008 American Association for Cancer Research.
`d0i:l0.l158/0008-5472.CAN-08-1776
`
`receptors. These receptors, which also include HER3 (ErbB3) and
`HER4 (ErbB4), are known to play critical roles in both development
`and cancer (1, 2). Importantly, amplification and overexpression of
`HER2 occur in 20% to 25% of human breast cancer and are
`
`predictive of poor clinical outcome (3, 4). Because of the role of
`HER2 in breast cancer pathogenesis and the accessibility of the
`extracellular portion of the receptor, HER2 was recognized as a
`potential candidate for targeted antibody therapy. The humanized
`HER2 antibody,
`trastuzumab (Herceptin), was approved by the
`Food and Drug Administration in 1998 for use in metastatic breast
`cancer and has subsequently shown clinical benefit when used, in
`combination with cytotoxic chemotherapy, as first-line or adjuvant
`therapy (5, 6). Importantly, trastuzumab improves overall survival
`in early breast cancer after chemotherapy compared with
`observation alone (6). Increased survival after only 2 years of
`follow-up is impressive in breast cancer. Tamoxifen is the only
`other breast cancer treatment that is reported to offer a survival
`benefit in this short-time period (6).
`Although the mechanisms for response to trastuzumab are not
`completely understood, clinical benefit is attributed to interference
`with signal transduction pathways,
`impairment of extracellular
`domain (ECD) cleavage,
`inhibition of DNA repair, decreased
`angiogenesis; as well as induction of cell cycle arrest, and
`antibody-mediated cellular cytotoxicity (7, 8). Despite these diverse
`mechanisms of action, a significant proportion of patients treated
`with trastuzumab either do not respond initially or relapse after
`experiencing a period of clinical response (5, 9). Progression
`through trastuzumab-containing therapy is attributed to aberrant
`activation of signaling pathways, such as the phosphatidylinositol
`3-kinase pathway (10-12), activation of compensatory signaling
`either through up-regulation of the insulin-like growth factor-I
`receptor (13, 14) or ErbB/HER ligands (15, 16) or generation of a
`constitutively active truncated form of HER2, designated p95HER2
`(17, 18).
`Direct covalent coupling of cytotoxic agents to monoclonal
`antibodies is an alternative to naked antibody-targeted therapy.
`To date, antitumor antibodies have been linked to cytotoxic agents,
`such as the calicheamicins, auristatins, maytansinoids and
`derivatives of CC1065 (19-22). Currently, only one such conjugate,
`anti-CD33 conjugated to calicheamicin (gemtuzumab ozogamicin
`or Mylotarg), has been granted marketing approval
`for
`the
`treatment of relapsed acute myeloid leukemia (23).
`Maytansinoids are derivatives of the antimitotic drug maytan-
`sine. These agents bind directly to microtubules in a manner
`similar to the Vinca alkaloids (24, 25). Antibody-maytansinoid
`conjugates directed toward cancer antigens,
`such as CanAg
`(cantuzumab mertansine and IMGN242), prostate-specific mem-
`brane antigen (MLN2704), CD56 (IMGN901), CD33 (AVE9633), and
`
`Cancer Res 2008; 68: (22). November 15, 2008
`
`9280
`
`www.aacrjourna|s.org
`
`Downloaded from cancerres.aacrjourna|s.org on February 9, 2014. © 2008 American Association for Cancer
`Research.
`
`PHIGENIX
`
`Exhibit 1004-02
`
`

`
`CD44v6 (bivatuzumab mertansine) are in early stages of clinical
`testing (20, 26-28). Because HER2 is highly differentially expressed
`on breast tumor cells (1-2 million copies per cell) compared with
`normal epithelial cells, HER2 represents an ideal
`target
`for
`antibody—drug conjugate (ADC)
`therapy. Numerous preclinical
`and clinical studies indicate that trastuzumab combines extremely
`well with microtubule-directed agents (29-32). Given the mecha-
`nism of action and potency of maytansine, it was deemed to be a
`particularly attractive cytotoxic agent to conjugate to trastuzumab.
`Herein, we describe the efficacy, pharmacokinetic properties, and
`safety of several
`trastuzumab—maytansinoid conjugates, with
`particular emphasis on the chemical nature of the linker.
`
`Materials and Methods
`
`Cell lines and reagents. Tumor cell lines (breast carcinoma BT—474, SK-
`BR—3, MCF7, MDA—MB—468, MDA—MB—361, HCC1954, lung carcinoma Calu 3,
`and ovarian carcinoma line SK—OV—3) and MCF 10A breast epithelial cells
`were obtained from American Type Culture Collection. The breast tumor
`line KPL—4 was obtained from Dr. J. Kurebayashi (33), and MKN7 gastric
`carcinoma cells were from Mitsubishi Corp. Cells were maintained in Ham’s
`F-12: high glucose DNEM (50:50) supplemented with 10% heat—inactivated
`fetal bovine serum and 2 mmol/L L—glutamine (all from Invitrogen Corp.).
`Normal human cell lines [human mammary epithelial cells (HMEC) and
`normal human epidermal keratinocytes (NHEK)] and the corresponding
`culture medium (MEGM and KGM,
`respectively) were obtained from
`Cambrex. The BT474—EEI cell line was derived by subculturing BT—474
`tumors grown in viva in the absence of estrogen pellet supplementation
`(exogenous estrogen independent) and is resistant both in Vitro and in Vii/0
`to trastuzumab treatment.
`
`Active agents used for cell culture and animal studies were the antibody
`trastuzumab (Genentech, Inc.), trastuzumab—maytansinoid ADC (Immuno-
`Gen, Inc.), and the control ADC, anti—H—8—MCC—DM1. The maytansinoid,
`DM1, was conjugated to trastuzumab through SPP, SMCC, or SPDP linkers
`(Fig. 1; refs. 24, 34); the thiol—containing maytansinoids, DM3 and DM4,
`
`Trastuzumab-DM1 Antibody-Drug Conjugate
`
`which have methyl groups adjacent to their sulfhydryl group were linked to
`trastuzumab with the SSNPP linker (ImmunoGen, Inc.). All trastuzumab
`ADCs had an average molar ratio of 3 to 3.6 maytansinoid molecules per
`antibody. The drug—antibody molar ratio for trastuzumab—MCC—DM1 and
`trastuzumab—SPP—DM1 was 3.2 for cell culture and xenograft studies, 3.6 for
`trastuzumab—SPP—DM1 used in the rat toxicity study, and 3.8 for anti—H—8—
`MCC—DM1.
`
`Cell viability and cell death assays. The effects of trastuzumab and
`trastuzumab—maytansinoid conjugates on tumor cell viability were assessed
`using Cell Titer—Glo (Promega Corp.). Cells were plated in black—walled
`96-well plates (20,000 per well for BT—474; 10,000 cells per well for all other
`lines) and allowed to adhere overnight at 37 °C in a humidified atmosphere
`of 5% CO2. Medium was then removed and replaced by fresh culture
`medium containing different concentrations of trastuzumab, trastuzumab
`ADC, or free DM1, and the cells incubated for varying periods of time. After
`each time point, Cell Titer—Glo reagent was added to the wells for 10 min at
`room temperature and the luminescent signal was measured using a
`Packard/Perkin—Elmer TopCount. For measurement of apoptosis, BT—474
`and SK—BR—3 were exposed to trastuzumab or trastuzumab—MCC—DM1 for
`48 h. Caspase activation was assessed by adding Caspase—Glo 3/7 reagent
`(Promega Corp.) for 30 min at room temperature, and the luminescence
`was recorded using a Packard TopCount Induction of cytotoxicity was
`assessed in cells treated with trastuzumab or trastuzumab—MCC—DM1 for
`
`72 h using ToxiLight Bioassay kit (Cambrex/Lonza). This assay measures
`release of the intracellular enzyme adenylate kinase as a result of cell lysis.
`Normal HMEC and NHEK were plated in clear 96-well plates at densities
`of 10,000 and 8,000 cells per well, respectively, and allowed to adhere
`overnight. Cells were treated with trastuzumab or trastuzumab—MCC—DM1
`for 72 h. Alamar Blue reagent (Trek Diagnostics Systems) was added to all
`wells, plates were incubated for 3 h at 37 °C, and fluorescence was measured
`on a SpectraMax 190 (Molecular Devices) using 530—nm excitation and
`590—nm emission. Because the normal cell lines were not healthy when
`grown in black multiplates (which is necessary for use of Cell Titer—Glo),
`Alamar Blue was used as the proliferation read—out. For all cellular assays,
`dose—response curves were generated using Kaleidagraph 4.0 (Synergy
`Software) four—parameter curve fitting.
`
`finker
`
`T:‘s,.siLl2umab {Tm 53)
`
`E’
`
`'l'rs3s‘1E3-Si-‘D5-7-DM1
`
`Figure 1. Structure of trastuzumab
`(Tmab)—maytansinoid conjugates (stability of
`linker, least to greatest): Tmab—SPDP—DM1,
`Tmab-SPP—DM1, Tmab—SSNPP—DM3,
`Tmab—SSNPP—DM4, and Tmab—SMCC-DM1
`(nonreducible).
`
`-
`
`Me
`
`Drug Maytansinoid
`(DM)
`
`Tamil}-SP?-DM1
`
`Trst.=;‘zE3-SSNPP-DM3
`
`'i'rn.=szt3-€~Z€~1E\E{JP-DM4
`
`f T:*n.aE3-MEL”,-C-DM1
`
`www.aacrjourna|s.org
`
`9281
`
`Cancer Res 2008; 68: (22). November 15, 2008
`
`Downloaded from cancerres.aacrjourna|s.org on February 9, 2014. © 2008 American Association for Cancer
`Research.
`
`PHIGENIX
`
`Exhibit 1004-03
`
`

`
`Cancer Research
`
`Western immunoblot analysis. SK—BR—3 cells were seeded at a density
`of 1 million per dish in 100 X 15 mm dishes and allowed to adhere for 2 d.
`The medium was then removed and replaced with fresh medium
`containing either trastuzumab, free DM1, or a range of concentrations of
`trastuzumab—MCC—DM1. After a 48-h incubation,
`floating cells were
`collected and combined with detached adherent cells. The total cell
`
`population was then centrifuged and resuspended in lysis buffer [50 mmol/L
`HEPES (pH 7.5), 150 mmol/L NaCl, 1.5 mmol/L MgCl2, 1.0 mmol/L EGTA,
`10% glycerol, 1% Triton X-100, 10 mmol/L Na4P2O4, 1 mmol/L Na3VO4,
`50 mmol/L NaF,
`1
`].l1’I1()l/L leupeptin, 0.3 ].l1’I1()l/L aprotinin,
`1
`].l1’I1()l/L
`pepstatin A, 10 iimol/L bestatin, and 1.4 ].l1’I10l/L E—64]. Lysates were cleared
`by centrifugation at 4°C for 15 min at 20,800 X g in a microcentrifuge, and
`protein concentrations were determined using the bicinchoninic acid (BCA)
`protein assay kit (Pierce). Proteins were resolved by SDS—PAGE, transferred
`to nitrocellulose, and immunoblotted with a polyclonal antibody against
`poly(ADP—ribose) polymerase (PARP), which recognizes intact 116—kDa PARP
`and the 23-kDa cleavage fragment (R&D Systems). Blotting was carried out
`in TBS containing 0.1% Triton X-100 and 5% nonfat dry milk, followed by
`incubation with horseradish peroxidase (HRP)—conjugated secondary anti-
`bodies (Amersham Biosciences). Proteins were visualized using enhanced
`chemiluminescence reagents (Amersham Biosciences).
`Measurement of total and phosphorylated HER2 and p95HER2 in
`transgenic tumors was performed as follows. Tumors from the founder 5
`(Fo5;
`ref. 35) and founder 2#1282 (F2#1282)
`lineages of MMTV—HER2
`transgenic mice (Genentech, Inc.) were excised from the animals, placed in
`lysis buffer containing protease inhibitors, and homogenized on ice. Tumor
`lysates were centrifuged, and total protein levels in the supernatant were
`determined using a BCA protein assay kit. HER2 was immunoprecipitated
`overnight at 4°C using the mouse monoclonal antibody Ab—15 (Lab Vision)
`complexed to protein A/G sepharose, with 1 mg total protein from at least
`three independent tumor lysates. Complexes were pelleted by centrifuga-
`tion, washed twice with lysis buffer, resuspended in SDS sample buffer, and
`boiled. Samples were separated on a 4% to 12% Tris—glycine gel and
`transferred to nitrocellulose membranes. Blots were probed with mouse
`monoclonal antibody Ab—18 (Lab Vision)
`to detect the phosphorylated
`forms of HER2 and p95HER2 or with Ab—15 to detect total HER2 and
`p95HER2.
`In viva efficacy and pharmacokinetic studies. Tumor tissue from
`Fo5 or F2#1282 HER2 transgenic mice was collected aseptically, rinsed in
`HBSS, and cut into pieces of ~2 X 2 mm in size. These pieces were
`surgically transplanted into the mammary fat pad of female nu/nu mice
`(Charles River Laboratories). For efficacy studies using BT474EEI cells,
`naive female beige nude XID mice (Harlan Sprague—Dawley) were
`inoculated in the mammary fat pad with 20 million tumor cells
`suspended in 50% phenol red—free Matrigel (Becton Dickinson Bioscience)
`mixed with culture medium. All animals were randomly assigned into
`treatment groups, such that the mean tumor volume for each group was
`100 to 200 mm°. Trastuzumab or trastuzumab—maytansinoid conjugates
`were given by either a single i.v. injection or injection once every 3 wk.
`Vehicle control was either PBS (for pharmacokinetic studies) or ADC
`formulation buffer
`[10 mmol/L sodium succinate, 0.1% polysorbate
`(Tween) 20, 20 mg/mL trehalose dihydrate (pH5.0)]. Similarly, KPL—4
`human breast tumor cells were inoculated (3 million cells per mouse, in
`Matrigel) into the mammary fat pads of SCID beige mice (Charles River
`Laboratories). Trastuzumab (15 mg/kg) was given i.p. once per week for
`4 wk; trastuzumab—MCC—DM1 (15 mg/kg) was given i.v. (single injection
`on treatment day 0). All treatment groups consisted of 6 to 10 animals per
`group, and tumor
`size was monitored twice weekly using caliper
`measurement. Mice were housed in standard rodent microisolator cages.
`Environmental controls for the animal rooms were set
`to maintain a
`
`temperature of ~70°F, a relative humidity of 40% to 60%, and an
`approximate 14-h light/10-h dark cycle.
`For pharmacokinetic analysis of trastuzumab—maytansinoid conjugates,
`female beige nude mice (age 15-20 wk; Harlan Sprague—Dawley) were
`injected i.v. with 2 mg/kg of different trastuzumab ADCs (four mice per
`group). To assess circulating levels of total and conjugated antibody, blood
`was collected via cardiac puncture from three animals at 5 min and 1, 6, 24,
`
`72, and 168 h postinjection. The samples were left at room temperature for
`30 min until the blood coagulated. Subsequently, serum was obtained by
`centrifuging the samples at 10,000 X g for 5 min at 4°C, after which serum
`samples were stored at —70°C. Total trastuzumab concentration in the
`serum samples was measured as follows: 96-well ELISA plates were coated
`with HER2 ECD in 0.05 mol/L carbonate/bicarbonate buffer (pH 9.6) at 4°C
`overnight. After removal of the coat solution, nonspecific binding sites were
`blocked by incubating with blocking solution [0.5% bovine serum albumin
`(BSA) in PBS] for 1 to 2 h. The plates were then washed with wash buffer
`(0.05% Tween in PBS), and standards or samples diluted in ELISA assay
`buffer [PBS containing 0.5% BSA, 0.05% Tween, 10 ppm proclin 300, 0.2%
`bovine 'y—globulin, 0.25% CHAPS, 0.35 mol/L NaCl, 5 mmol/L EDTA (pH 7.4)]
`were added. After a 2—h
`incubation, plates were washed and HRP-
`conjugated goat anti—human Fc (The Jackson Laboratory) was added for
`an additional 2 h. Plates were then washed again, followed by the addition
`of tetramethyl benzidine substrate for color development. The reaction was
`stopped after 10 to 15 min by the addition of 1 mol/L phosphoric acid.
`Plates were read on a Molecular Devices microplate reader at a wavelength
`of 450 to 630 nm. The concentration of trastuzumab in the samples was
`extrapolated from a four—variable fit of the standard curve. For
`measurement of trastuzumab—maytansinoid concentration, wells were
`coated with anti—DM antibody (GNE DM1—3586) and serum samples
`added as above. After the 2—h sample incubation, the plates were washed,
`60 ng/mL biotin—conjugated HER2 ECD was added to each well, and the
`plates were incubated for
`1 h. Plates were then washed, and HRP-
`conjugated streptavidin (GE Healthcare) was added for an additional
`30-min incubation. Color detection and measurement were performed as
`described above. Previous analyses of different preparations of trastuzu-
`mab—DM1 conjugates with drug—antibody ratios ranging from 1.9 to 3.8
`showed that the conjugated antibody ELISA is not sensitive to drug load.
`Circulating HER2 ECD levels from mice harboring Fo5 or F2#1282
`xenograft tumors was measured, as previously described (35). Briefly, serum
`was diluted 1:50 with ELISA assay buffer (above) followed by 1:2 serial
`dilutions. HER2 ECD was captured using goat anti—HER2 polyclonal
`antibody (Genentech,
`Inc.) and detected with biotin—conjugated rabbit
`anti—Her2 polyclonal (Genentech, Inc.), followed by addition of streptavidin-
`HRP.
`
`Rat toxicity studies. Female Sprague—Dawley rats weighing 75 to 80 g
`were obtained from Charles River Laboratories and allowed to acclimate
`
`trastuzumab—MCC—DM1,
`for 5 d before study. Trastuzumab—SPP—DM1,
`and free DM1 were diluted in PBS (Invitrogen Corp.) as vehicle and given
`as a single i.v. bolus tail—vein injection on day 1 at a dose volume of
`10 mL/kg. Body weights were measured predose on day 1 and daily
`thereafter for 5 d.
`
`Results
`
`Linker optimization. Antibody—DM1 conjugates were originally
`designed with a disulfide-based linker for release of active drug by
`intracellular reduction (24). Recently, it was discovered that the
`endocytic pathway is oxidizing and that cleavage of the disulfide
`linker, SPP,
`is very inefficient
`(36). Thus, different trastuzumab
`ADCS were constructed to investigate the effect of disulfide linker
`hindrance on the biological activity of these conjugates (Fig. 1). A
`trastuzurnab—DM1 conjugate made with the SPDP linker contains
`no methyl substitutions adjacent to the disulfide bond and is
`therefore the least hindered disulfide—containing design. Trastuzu-
`mab ADCS composed of SPP—DM1, SSNPP—DM3, or SSNPP-DM4
`contain one, two, or three methyl groups, respectively, around the
`disulfide bridge and show increasing resistance to cleavage via
`thiol-disulfide exchange reactions?’ DMS and DM4 nomenclature
`
`3 ImmunoGen, Inc., unpublished data.
`
`Cancer Res 2008; 68: (22). November 15, 2008
`
`9282
`
`www.aacrjourna|s.org
`
`Downloaded from cancerres.aacrjourna|s.org on February 9, 2014. © 2008 American Association for Cancer
`Research.
`
`PHIGENIX
`
`Exhibit 1004-04
`
`

`
`reflects addition of methyl groups to the DM1 (drug) moiety
`(addition of one or two methyls, respectively). An additional ADC
`containing a thioether linker
`(SMCC, designated MCC after
`conjugation) was also constructed. In Vitro potency assays were
`conducted for 3 days with the HER2-amplified breast cancer lines
`BT-474 and SK-BR-3 treated with various trastuzumab ADCs. This
`
`short treatment period was selected to minimize the effects of
`unconjugated trastuzumab on these trastuzumab-sensitive lines.
`The graphs in Fig. 2A show enhanced potency of trastuzumab-
`maytansinoid conjugates compared with trastuzumab in both cell
`lines, with no significant difference in activity among the various
`ADCs tested (IC50, 0.085—0.148 ug/mL for BT-474 and 0.007-
`0.018 ug/mL for SK-BR-3). Because the effect of trastuzumab is
`cytostatic in nature, the enhanced potency of the ADC is, thus, due
`to exposure of the cells to the cytotoxic maytansinoid. In addition,
`brief exposure of SK-BR-3 cells for 10, 30, or 60 minutes to
`trastuzumab-MCC-DM1, followed by a 3-day incubation in culture
`medium, also resulted in substantial growth inhibition (data not
`shown). When comparing potency as molar DM1 equivalents, DM1
`conjugated to trastuzumab is 5-fold more potent than free L-DM1
`on SK-BR-3 and shows equal potency to free L-DM1 on BT-474 cells
`(Fig. 2B). Nontargeted effects of trastuzumab-maytansinoid con-
`jugates were assessed on breast cancer lines expressing normal
`levels (MCF7) or lacking expression (MDA-MB-468) of HER2 (37).
`All ADCs tested showed minimal antiproliferative activity in both
`cell lines (Fig. 2A), whereas free DM1 showed potency equal to that
`observed on the HER2-amplified lines (Fig. 2B). Treatment of the
`four cell lines with an isotype-matched control ADC, anti—IL-8-
`MCC-DM1,
`resulted in negligible growth inhibition (data not
`shown). Thus, like trastuzumab, maytansinoid-conjugated trastu-
`zumab ADCs are specific for HER2-overexpressing cells. Further-
`more, the different linkers behave similarly with respect to in Vitro
`antiproliferative activity.
`Pharmacokinetic analyses in nude mice were performed to
`determine the effect of different
`trastuzumab ADC linkers on
`serum concentration.
`In contrast
`to the cell culture results,
`we observed a clear correlation between ADC exposure and
`linker hindrance. The ADC with the least hindered disulfide bond
`
`(no adjacent methyl groups), trastuzumab-SPDP-DM1, showed the
`fastest clearance and was undetectable by day 3. The addition of
`methyl groups (CH3) on either side of the S-S bond (trastuzumab-
`SPP-DM1 with one CH3 on the antibody side of the S-S;
`trastuzumab-SSNPP-DM3 with two CH3, one on either side of
`the S-S;
`trastuzumab-SSNPP-DM4 with three CH3, one on the
`antibody side, and two on the drug side of the S-S) resulted in
`increasingly sustained ADC serum concentrations (Fig. 3A,
`left).
`Trastuzumab-MCC-DM1 (nonreducible) and trastuzumab-SSNPP-
`DM4 (the most hindered disulfide-containing design) showed
`similar pharmacokinetics with the ADC concentration at 70% of
`the total antibody at day 7. Studies were also performed to
`compare clearance of DM1-conjugated trastuzumab to total
`trastuzumab levels (Fig. 3A,
`right). Serum concentrations of
`trastuzumab-MCC-DM1 measured for 1 week were not different
`
`from total serum trastuzumab concentration, indicating that, with
`the nonreducible MCC linker,
`the amount of maytansinoid
`released from the antibody was negligible over the 7 days. In
`contrast, only 11% of the trastuzumab-SPP-DM1, compared with
`total
`trastuzumab antibody concentration,
`remained in the
`circulation after 7 days.
`In Vivo efficacy studies examining the different trastuzumab-
`maytansinoid conjugates given as a single i.v. dose were carried
`
`Trastuzumab-DM1 Antibody-Drug Conjugate
`
`out using the MMTV-HER2 Fo5 mammary tumor transplant
`(trastuzumab-resistant) model. This model was developed by
`serial
`transplantation of tumors derived from the HER2
`transgenic mouse lineage founder 5 (35). These mouse tumors
`overexpress human HER2 (3+ expression) as measured by
`immunohistochemistry and quantitative reverse transcription-
`PCR. Interestingly, established tumors from this model do not
`respond to single-agent trastuzumab therapy. Consistent with the
`pharmacokinetic analysis,
`increased linker
`stability in viva
`correlated with increased antitumor activity for
`trastuzumab
`ADCs (Fig. 3B). Tumor volume in both trastuzumab-MCC-DM1
`and trastuzumab-SSNPP-DM3 treatment groups were statistically
`different from trastuzumab-SPP-DM1 (log-rank P values of 0.0165
`and 0.0414, respectively). Further characterization of the HER2
`transgenic models revealed the presence of high circulating levels
`of shed HER2 ECD in the serum of Fo5 tumor-bearing mice.
`Moreover, analysis of these tumors revealed increased expression
`of the transmembrane-containing fragment p95HER2, which is
`highly tyrosine-phosphorylated, indicating constitutive activation.
`In comparison, the trastuzumab-sensitive F2#1282 model showed
`1,000-fold lower levels of circulating HER2 ECD, no detectable
`p95HER2, and significant tumor growth reduction after a single
`injection of different doses of trastuzumab-DM1 (data not
`shown). Thus, the trastuzumab-DM1 conjugate is efficacious in
`models,
`such as Fo5, which express
`reported markers of
`resistance to trastuzumab,
`i.e., high circulating HER2 ECD and
`activated p95HER2.
`Short-term,
`single-dose toxicity studies were performed in
`female rats comparing the effects on body weight of free or
`conjugated (SMCC or SPP) DM1. ADCs were given as DM1
`equivalents (Hg/m2). As
`trastuzumab does not
`recognize rat
`erbB2, this model measures antigen-independent ADC effects. Of
`the agents tested, trastuzumab-MCC-DM1 showed the best safety
`profile. Body weight gain in rats given 1,632 ug/m2 DM1 (25 mg/
`kg antibody) was comparable with vehicle control animals (15.9%
`and 16.3%, respectively; Fig. 3C). In contrast, administration of
`trastuzumab-SPP-DM1 at an equivalent DM1 dose resulted in
`considerable body weight
`loss (—10%) by end of study. Rats
`treated with 3,264 Hg/m2 (50 mg/kg) trastuzumab-MCC-DM1 did
`not exhibit body weight
`loss; however,
`the amount of weight
`gained during the course of study (+6.7%) was less than in the
`vehicle control and the 1,632 ug/m2 trastuzumab-MCC-DM1
`groups. The change in body weight with 3,264 ug/m2 trastuzu-
`mab-MCC-DM1 was similar to that observed with free DM1 at a
`
`dose of 653 Hg/m2 (+6.5%). In a separate study, clinical chemistry
`analysis of rats treated with trastuzumab-MCC-DM1 showed
`transient elevation of liver enzymes (aspartate aminotransferase,
`alanine aminotransferase, and ~/-glutamyl
`transpeptidase) and
`mild, reversible thrombocytopenia at a dose of 20 mg/kg, and no
`clinical signs of toxicity at 6 mg/kg (data not shown). Evidently,
`substituting a reducible linker (SPP) with a nonreducible linker
`(SMCC) results in a less toxic but still efficacious trastuzumab-
`DM1 conjugate. Consequently, fiirther studies focused on defining
`the activity of trastuzumab-MCC-DM1.
`tumor cells
`HER2-overexpressing,
`trastuzumab-resistant
`respond to trastuzumab-MCC-DM1, whereas normal cell lines
`are unaffected. The effects of trastuzumab-MCC-DM1 on in Vitro
`
`proliferation were fiirther examined in several tumor lines shown
`to be resistant to trastuzumab. Additional breast cancer lines,
`HCC1954, KPL-4, and BT-474 EEI, all with 3+ HER2 expression,
`were first
`tested. The BT-474 EEI cell
`line was developed by
`
`www.aacrjourna|s.org
`
`9283
`
`Cancer Res 2008; 68: (22). November 15, 2008
`
`Downloaded from cancerres.aacrjourna|s.org on February 9, 2014. © 2008 American Association for Cancer
`Research.
`
`PHIGENIX
`
`Exhibit 1004-05
`
`

`
`Cancer Research
`
`
`Relativecell
`
`
`
`viability(cellularATPcontent,percentofcontrol)
`
`>3:
`".5
`E5>
`T;L)
`cu
`.2..
`‘.5an
`
`
`
`
`
`LC(cellularATPcontent,percentofcontrol)
`
`Relativecell
`
`
`
`
`
`viability(cellularATPcontent,percentofcontrol)
`
`0.001
`
`0.01
`
`0.1
`
`1
`
`Trastuzumab-DM concentration (ug/mL)
`
`;mS.-”~‘F‘mD:'\/it
`
`ii‘.*a2ii:i~5.§§.‘>N§-‘t-‘~L:
`
`
`Relativecell
`
`
`
`viability(cellularATPcontent,percentofcontrol)
`
`00O
`
`-l>O
`
`|\JCD0O
`
`00O
`
`0.001
`
`0.01
`
`0.1
`
`1
`
`Trastuzumab-DM concentration (ug/mL)
`
`MDA-MB-468
`
`0.001
`
`0.01
`
`0.1
`
`1
`
`0.001
`
`0.01
`
`0.1
`
`1
`
`Trastuzumab-DM concentration (pg/mL)
`
`Trastuzumab-DM concentration (pg/mL)
`
`In vitro activity of trastuzumab-DM1 conjugates versus free DM1
`
`(K250, nmol/L DM1 equivalents)
`
`trastuzumab-SPF’-DM1
`
`trastuzumab-MCC-DM1
`
`free L-DM1
`
`SK-BR-3
`
`BT-474
`
`MCF7
`
`MDA-MB-468
`
`0.24
`
`2.71
`
`>100
`
`18.37
`
`0.22
`
`4.26
`
`>100
`
`41.37
`
`1.00
`
`4.52
`
`2.92
`
`0.59
`
`Cells were treated with trastuzumab-DM1 conjugates or L-DM1 as free drug for72 h;
`|C50 values were determined from four-parameter curve fitting.
`
`Figure 2. Trastuzumab—maytansinoid conjugates are selective for HER2—amp|ified breast cancer lines, whereas free DM1 shows equivalent potency on all cell lines
`tested regardless of HER2 expression. A, trastuzumab—maytansinoid conjugates show enhanced antiproliferative activity on HER2—overexpressing breast tumor cell
`lines compared with trastuzumab. The nature of the linker, however, does not affect in vitro activity. B, in vitro potency of Tmab—SPP—DM1 and Tmab—MCC—DM1
`compared with the free drug L-DM1 afler 3 d of treatment (lC50 values, nmol/L DM1 equivalents). HER2 expression: SK—BR—3 (HER2 3+), BT-474 (HER2 3+), MCF7
`(normal HER2 expression), MDA-MB-468 (HER2—negative).
`
`Cancer Res 2008; 68: (22). November 15, 2008
`
`9284
`
`www.aacrjourna|s.org
`
`Downloaded from cancerres.aacrjournals.org on February 9, 2014. © 2008 American Association for Cancer
`Research.
`
`PHIGENIX
`
`Exhibit 1004-06
`
`

`
`Trastuzumab-DM1 Antibody-Drug Conjugate
`
`If
`E\
`cs:
`3C
`.9..
`cu
`-.5C
`0:C.)C
`
`oDE3.
`
`_
`as
`CD
`
`DM1-conjugatedantibody(%oftotal
`
`antibody)
`
`‘M.-.'-.
`
`-.5.”
`
`’3‘i':':£.‘ii::f-J5“.-‘('";?€§"‘§*"*3'.'?i“‘fi3
`
`00O0
`
`ETAO0
`
`
`
`Percentweightchange
`
`
`
`(meaniSE)
`
`
`
`Meantumorvolume(mm
`
`
`
`
`
`Figure 3. Trastuzumab conjugated to DM1 through the thioether SMCC linker displays better efficacy and pharmacokinetics and lower toxicity than conjugates
`with disulfide linkers. A, pharmacokinetic analysis of trastuzumab—maytansinoid conjugates shows increased serum concentrations of conjugates with more hindered
`disulfide linkers. Female beige nude mice were given a single i.v. injection (2 mg/kg) of different trastuzumab—maytansinoid conjugates (n = 4 mice per group).
`Serum samples were collected at various time points afler injection for measurement of total and conjugated trastuzumab as described. B, increased in vivo linker
`stability results in improved efficacy in vivo. Mice bearing mammary tumor transplants from the MMTV—HER2 Fo5 line were given a single i.v. injection (10 mg/kg)
`of Tmab—SPP—DM1, Tmab—SSNPP—DM3, Tmab—SSNPP—DM4, Tmab—MCC—DM1, or vehicle (n = 7 mice per group), and tumor growth was monitored for 25 d.
`C, Tmab—MCC—DM1 displays the best safety profile, as assessed by changes in body weight, of the conjugates tested. Female Sprague—Dawley rats were given a
`single i.v. injection of Tmab—MCC—DM1 (1,632 or 3,264 pg/m2, 25 or 50 mg/kg, respectively), Tmab—SPP—DM1 (1,6

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