throbber
Published OnlineFirst March 14, 2013; DOI: 10.1158/1535-7163.MCT-13-0002
`
`Molecular
`Cancer
`Therapeutics
`
`Large Molecule Therapeutics
`
`Construction and Characterization of Novel, Completely
`Human Serine Protease Therapeutics Targeting Her2/neu
`
`Yu Cao, Khalid A. Mohamedali, John W. Marks, Lawrence H. Cheung, Walter N. Hittelman, and
`Michael G. Rosenblum
`
`Abstract
`
`Immunotoxins containing bacterial or plant toxins have shown promise in cancer-targeted therapy, but
`their long-term clinical use may be hampered by vascular leak syndrome and immunogenicity of the
`toxin. We incorporated human granzyme B (GrB) as an effector and generated completely human
`chimeric fusion proteins containing the humanized anti-Her2/neu single-chain antibody 4D5 (designated
`GrB/4D5). Introduction of a pH-sensitive fusogenic peptide (designated GrB/4D5/26) resulted in
`comparatively greater specific cytotoxicity although both constructs showed similar affinity to Her2/
`neu–positive tumor cells. Compared with GrB/4D5, GrB/4D5/26 showed enhanced and long-lasting
`cellular uptake and improved delivery of GrB to the cytosol of target cells. Treatment with nanomolar
`concentrations of GrB/4D5/26 resulted in specific cytotoxicity, induction of apoptosis, and efficient
`downregulation of PI3K/Akt and Ras/ERK pathways. The endogenous presence of the GrB proteinase
`inhibitor 9 did not impact the response of cells to the fusion construct. Surprisingly, tumor cells resistant
`to lapatinib or Herceptin, and cells expressing MDR-1 resistant to chemotherapeutic agents showed no
`cross-resistance to the GrB-based fusion proteins. Administration (intravenous, tail vein) of GrB/4D5/26
`to mice bearing BT474 M1 breast tumors resulted in significant tumor suppression. In addition, tumor
`tissue excised from GrB/4D5/26–treated mice showed excellent delivery of GrB to tumors and a dramatic
`induction of apoptosis compared with saline treatment. This study clearly showed that the completely
`human, functionalized GrB construct can effectively target Her2/neu–expressing cells and displays
`impressive in vitro and in vivo activity. This construct should be evaluated further for clinical use.
`Mol Cancer Ther; 12(6); 979–91. Ó2013 AACR.
`
`Introduction
`Bacterial and plant toxin-based immunotoxins have
`shown remarkable potency and specificity, but a number
`of obstacles limit their clinical application (1, 2). The toxin
`component of these fusion proteins can elicit vascular
`damage leading to loss of vascular integrity (vascular leak
`syndrome, VLS; refs. 3, 4). Immune responses to the toxins
`in patients also result in rapid clearance of subsequent
`courses of therapy (5, 6). Toxin immunogenicity is being
`addressed by engineering B-cell epitopes on the structure
`
`Authors' Affiliation:
`Immunopharmacology and Targeted Therapy Lab-
`oratory, Department of Experimental Therapeutics, MD Anderson Cancer
`Center, Houston, Texas
`
`Note: Supplementary data for this article are available at Molecular Cancer
`Therapeutics Online (http://mct.aacrjournals.org/).
`
`Current address for Y. Cao: Department of Chemistry, The Scripps
`Research Institute, La Jolla, CA 92037.
`
`Corresponding Author: Michael G. Rosenblum, Department of Experi-
`mental Therapeutics, MD Anderson Cancer Center, Houston, TX 77054.
`Phone: 713-792-3554; Fax: 713-794-4261; E-mail:
`mrosenbl@mdanderson.org
`
`doi: 10.1158/1535-7163.MCT-13-0002
`Ó2013 American Association for Cancer Research.
`
`(7, 8), but these molecules may be difficult to humanize
`completely (9).
`A new class of immunotoxins have recently been devel-
`oped containing cytotoxic human proteins (10, 11). Gran-
`zyme B (GrB) is a well-known serine protease generated
`by cytotoxic lymphocytes to induce apoptotic cell death in
`target cells (12, 13). Our group first showed that various
`fusion constructs targeting tumor cells and tumor endo-
`thelium and containing GrB have impressive proapopto-
`tic and cytotoxic activity (14–18). Several other groups
`have since confirmed these findings using other GrB-
`containing constructs (19, 20). Because endogenous GrB
`is present in plasma in both normal and pathologic states,
`it is unlikely that this molecule would engender an
`immune response.
`Dalken and colleagues have described a GrB/FRP5
`fusion construct targeting Her2/neu, which displayed
`selective and rapid tumor cell killing in vitro (21). How-
`ever, studies have shown that
`the fusion construct
`required the presence of the endosome-disrupting agent
`chloroquine for biologic activity and suggested that an
`endosomal release process may be necessary for Her2/
`neu–targeted agents. Studies by Wang and colleagues
`suggested that incorporation of a furin-sensitive linker
`into GrB-based fusion constructs may promote effective
`
`www.aacrjournals.org
`
`979
`
`Downloaded from
`
`mct.aacrjournals.org
`
`on July 26, 2014. © 2013 American Association for Cancer Research.
`
`IMMUNOGEN 2015, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst March 14, 2013; DOI: 10.1158/1535-7163.MCT-13-0002
`
`Cao et al.
`
`cytoplasmic delivery of an active GrB fragment into target
`cells (22). However, the recombinant molecule seemed to
`be stable only when generated in situ by protein-expres-
`sing transfected cells.
`We previously examined a series of anti-Her2/neu
`single-chain antibodies (scFv) fused to the recombinant
`gelonin (rGel) toxin, and clearly showed that scFvs with
`11 mol/L) as opposed to high
`intermediate affinity (K
`d 10
`12 mol/L) were optimal carriers of protein
`affinity (K
`d 10
`toxins (23, 24). Therefore, we used an intermediate-affin-
`ity, humanized anti-Her2/neu scFv-designated 4D5 for
`the construction of our GrB-containing fusion constructs.
`In this study, we provided data on the cytotoxicity of
`Her2/neu–targeted GrB fusions against a panel of human
`tumor cell lines and explored the mechanism of in vitro
`activity of these fusion constructs. Finally, we showed the
`in vivo antitumor efficacy of the functionalized GrB chi-
`meric protein against a human breast xenograft model.
`
`Materials and Methods
`Cell lines
`The cell lines BT474 M1, NCI-N87, Calu3, MDA MB435,
`and Me180 were all obtained from American Type Cul-
`ture Collection (ATCC). The human breast cancer cell
`lines MDA MB453 and eB-1 were generously supplied
`by Drs. Zhen Fan and Dihua Yu (MD Anderson Cancer
`Center, Houston, TX). The BT474 M1 Herceptin- and
`lapatinib-resistant cells were derived from BT474 M1 cells
`after a 12-month selection in the continuous presence of 1
`mmol/L Herceptin or 1.5 mmol/L lapatinib. BT474 M1
`MDR-1 cells were generated by the transfection of plas-
`mid pHaMDR1 to parental BT474 M1 cells. The HEK 293T
`cell line was supplied by Dr. Bryant G. Darnay (MD
`Anderson Cancer Center). All cell lines were maintained
`in Dulbecco’s Modified Eagle Medium or RPMI-1640
`medium supplemented with 10% heat-inactivated FBS,
`2 mmol/L L-glutamine, and 1 mmol/L antibiotics.
`Cell lines were validated by short tandem repeat
`(STR) DNA fingerprinting using the AmpF‘STR Identi-
`filer Kit according to the manufacturer’s instructions
`(Applied Biosystems). The STR profiles were compared
`with known ATCC fingerprints (ATCC.org),
`to the
`Cell Line Integrated Molecular Authentication database
`(CLIMA) version 0.1.200808 (http://bioinformatics.
`istge.it/clima/; Nucleic Acids Research 37:D925-D932
`PMCID: PMC2686526) and to the MD Anderson finger-
`print database. The STR profiles matched known DNA
`fingerprints or were unique.
`
`Plasmid construction
`The GrB/4D5/26, GrB/4D5, GrB/26, and GrB DNA
`constructs were generated by an overlapping PCR meth-
`od. Illustrations of the constructs are shown in Fig. 1A. We
`designed a universal 218 linker (GSTSGSGKPGSGEG-
`STKG) incorporated between the individual components
`of GrB, 4D5, or peptide 26. Peptide 26 (AALEALAEA-
`LEALAEALEALAEAAAA) was generated from the 29-
`residue amphipathic peptide without the 3 C-terminal
`
`A
`
`GrB/4D5
`
`GrB
`
`GrB/4D5/26
`
`GrB
`
`26
`
`A.A.
`502
`546
`
`M.W.
`53932.4
`57900.6
`
`P.I.
`9.56
`9.23
`
`GrB/4D5
`GrB/4D5/26
`
`B
`
`kDa
`150
`100
`75
`
`50
`
`35
`
`25
`
`Nonreduced
`
`Reduced
`
`Figure 1. Construction and preparation of GrB-based fusion constructs.
`A, schematic diagram of GrB fusion constructs containing scFv 4D5 and
`GrB without or with fusogenic peptide 26. B, purified GrB-based chimeric
`proteins were analyzed by SDS–PAGE under nonreducing conditions.
`
`amino acids, which are responsible for dimerization (25).
`All construct genes were cloned into the mammalian cell
`expression vector pSecTag (Life Technologies).
`
`Expression, purification, and activation of GrB-based
`proteins
`The GrB-based proteins were expressed in HEK
`293T host cells and purified by immobilized metal
`affinity chromatography as described in Supplementary
`Methods.
`
`Determination of K
`d by ELISA
`The K
`d value and specificity of GrB-based protein
`samples were evaluated by ELISA on Her2/neu extracel-
`lular domain (ECD), Her2/neu-positive BT474 M1 cells,
`and Her2/neu–negative Me180 cells. Rabbit anti-c-myc
`antibody and horseradish peroxidase–conjugated goat
`anti-rabbit immunoglobulin G were used as tracers in
`this assay, as described previously (24).
`
`GrB activity assays
`The enzymatic activity of the GrB component was
`determined in a continuous colorimetric assay using
`N-a-t-butoxycarbonyl-L-alanyl-L-alanyl-L-aspartyl-thio-
`benzylester (BAADT) as a specific substrate (18). Assays
`consisted of commercial human GrB (Enzyme Systems
`
`980
`
`Mol Cancer Ther; 12(6) June 2013
`
`Molecular Cancer Therapeutics
`
`Downloaded from
`
`mct.aacrjournals.org
`
`on July 26, 2014. © 2013 American Association for Cancer Research.
`
`IMMUNOGEN 2015, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst March 14, 2013; DOI: 10.1158/1535-7163.MCT-13-0002
`
`Products) or GrB-based fusion proteins in BAADT at
`
`C. The change in absorbance at 405 nm was measured
`25
`on a Thermomax plate reader. Increases in sample absor-
`bance were converted to enzymatic rates by using an
`1 (mol/L)
`1 at 405 nm.
`extinction coefficient of 13,100 cm
`The specific activity of GrB-based fusion proteins was
`calculated using native GrB as the standard.
`
`Internalization analysis
`Immunofluorescence-based internalization studies
`were conducted using BT474 M1 and Me180 cells. Cells
`were treated with 25 nmol/L GrB/4D5/26 for 4 hours
`and subjected to immunofluorescent staining with anti-
`GrB antibody [fluorescein isothiocyanate (FITC)-conju-
`gated secondary antibody]. Nuclei were counterstained
`with propidium iodide (PI). Visualization of immuno-
`fluorescence was conducted with a Zeiss LSM510 con-
`focal laser scanning microscope Zeiss LSM510 (Carl
`Zeiss).
`
`In vitro cytotoxicity assays
`Log-phase cells were seeded (5  103/well) in 96-well
`plates and allowed to attach overnight. Cells were further
`incubated with various concentrations of GrB-based
`
`fusion proteins, GrB, or medium at 37
`C for 72 hours.
`Cell viability was determined using the crystal violet
`staining method as described previously (23).
`
`Annexin V/PI staining
`The Annexin V/PI staining assay was used to quanti-
`tatively determine the percentage of cells undergoing
`apoptosis after exposure to GrB/4D5/26. Cells were seed-
`ed onto 6-well plates (5  105 cells/ well) and incubated
`
`with 100 nmol/L GrB/4D5/26 at 37
`C for 24 or 48 hours.
`Aliquots of cells were washed with PBS and then incu-
`bated with Annexin V–FITC antibody. PI solution was
`added at the end of the incubation, and the cells were
`analyzed immediately by flow cytometry.
`
`Cytochrome c release assay and Bax translocation
`After treatment with GrB/4D5 or GrB/4D5/26, cells
`were collected and resuspended with 0.5 mL of 1
`cytosol extraction buffer mix (BioVision) and then
`homogenized in an ice-cold glass homogenizer. The
`homogenate was centrifuged, and the supernatant was
`collected and labeled as the cytosolic fraction. The
`pellet was resuspended in 0.1 mL of mitochondrial
`extraction buffer and saved as the mitochondrial
`fraction. Aliquots of each cytosolic and mitochondrial
`fraction were analyzed by Western blotting with anti-
`bodies recognizing cytochrome c and Bax (Santa Cruz
`Biotechnology).
`
`Assays for caspase activation and apoptosis
`Western blot analysis was used to identify activation of
`caspases-3 and -9 as well as PARP cleavage. In addition,
`apoptosis was analyzed using antibodies recognizing Bcl-
`2 and BID (Santa Cruz Biotechnology).
`
`Novel Functionalized GrB-Targeting Her2/neu
`
`Impact on cell signaling pathways
`After treatment, cell lysates were analyzed by Western
`blotting with antibodies recognizing Her2/neu and phos-
`phorylated (p)-mTOR (S2448; Cell Signaling Technology)
`as well as p-Her2/neu (Tyr877), p-Her2/neu (Tyr 1221/
`1222), EGF receptor, p-EGF receptor (Thr845), Her3, p-
`Her3 (Tyr1328), IGF1 receptor, p-IGF1 receptor (Tyr 1165/
`1166), estrogen receptor (ER), progesterone receptor (PR),
`Akt, p-Akt, extracellular signal–regulated kinase (ERK),
`p-ERK (Thr 177/Thr 160), PTEN, proteinase inhibitor 9
`(PI-9), and b-actin (all from Santa Cruz Biotechonology).
`Immunoreactive proteins were visualized by enhanced
`chemiluminescence.
`
`In vivo efficacy studies
`We used Balb/c nude mice to evaluate the in vivo effect
`of GrB/4D5/26 against aggressive breast cancer. Each
`mouse received a weekly subcutaneous injection of 3
`mg/kg estradiol cypoinate (26) starting 2 weeks before
`the injection of 1  107 BT474 M1 cells into the right flank.
`On the third day after cell inoculation, mice were injected
`intravenously (tail vein) either with saline or GrB/4D5/26
`(44 mg/kg) 5 times per week for 2 weeks. Animals were
`monitored and tumors were measured (calipers) for an
`additional 50 days.
`
`Immunofluorescence analysis
`Twenty-four hours after the final injection of saline or
`GrB/4D5/26, mice were sacrificed and tumor samples
`were frozen immediately in preparation for section
`slides. The sample slides were incubated with either
`anti-GrB antibody (FITC-conjugated secondary anti-
`body) or a terminal deoxynucleotidyl transferase–medi-
`ated dUTP nick end labeling (TUNEL) reaction mixture,
`as well as with an anti-mouse CD31 antibody (phyco-
`erythrin-conjugated secondary antibody), and were fur-
`ther subjected to nuclear counterstaining with Hoechst
`33342. Immunofluorescence observation was conducted
`under a Zeiss Axioplan 2 imaging microscope (Carl
`Zeiss).
`
`Results
`Construction, expression, and purification of GrB-
`based fusions
`The sequence of the humanized anti-Her2/neu scFv
`4D5 was derived from the published Herceptin light-
`and heavy-chain variable domain sequences (27). Pre-
`vious observations suggested that use of fusogenic
`peptides facilitates endosomal escape and delivery of
`large molecules into the cytotol (28, 29). We therefore
`incorporated the fusogenic peptide 26 (Fig. 1A). GrB-
`based fusions were generated by fusing GrB to 4D5
`with (designated GrB/4D5/26) or without (designated
`GrB/4D5) the addition of pH-sensitive fusogenic pep-
`tide 26 (AALEALAEALEALAEALEALAEAAAA)
`to
`the C-terminal of the construct. Furthermore, GrB and
`GrB/26 were used as controls. All fusion proteins were
`expressed in human embryonic kidney cells (HEK
`
`www.aacrjournals.org
`
`Mol Cancer Ther; 12(6) June 2013
`
`981
`
`Downloaded from
`
`mct.aacrjournals.org
`
`on July 26, 2014. © 2013 American Association for Cancer Research.
`
`IMMUNOGEN 2015, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst March 14, 2013; DOI: 10.1158/1535-7163.MCT-13-0002
`
`Cao et al.
`
`293T). Following purification, the final products migrat-
`ed at the expected molecular weights, with a purity of
`more than 95% (Fig. 1B).
`
`Analysis of binding affinity
`The binding affinities (K
`d values) of GrB/4D5/26 and
`GrB/4D5 were assessed by ELISA using purified Her2/
`neu ECD, Her2/neu–positive BT474 M1 human breast
`cancer cells, and Her2/neu–negative Me180 human cer-
`vical cancer cells. Both fusions specifically bound to Her2/
`neu ECD and BT474 M1 cells but not to Me180 cells (Fig.
`2A). The apparent K
`d values were determined by calcu-
`lating the concentration of fusion constructs that pro-
`duced half-maximal specific binding. GrB/4D5 and
`GrB/4D5/26 showed apparent K
`d values of 0.329 and
`0.469 nmol/L, respectively, to Her2/neu ECD and 0.383
`and 0.655 nmol/L, respectively, to BT474 M1 cells. These
`results are in general agreement with the published K
`d
`value for native Herceptin to the Her2/neu receptor (0.15
`nmol/L; ref. 27).
`
`Enzymatic assay of GrB-based fusions
`To assess the biologic activity of the GrB component of
`the fusions, we compared the ability of the constructs to
`cleave the substrate BAADT with that of native, authentic
`GrB (Fig. 2B). GrB/4D5 and GrB/4D5/26 had intact GrB
`enzymatic activity (1.54  105 and 1.57  105 U/mmoL,
`respectively). These activities were comparable with that
`of the native GrB standard (1.19  105 U/mmoL). Because
`the pro-GrB fusion constructs contain purification tags on
`the N-terminal end of GrB and render the molecule
`enzymatically inactive, these proteins were unable to
`cause hydrolysis of BAADT.
`
`Cellular uptake and GrB delivery of fusion constructs
`Immunofluorescence staining was conducted with
`BT474 M1 and Me180 cells. The GrB moiety of both fusions
`was observed primarily in the cytosol after treatment with
`a fusion protein in BT474 M1 cells but not in Me180 cells
`(Fig. 2C), showing that both constructs were efficient in
`cell binding and internalization after exposure to Her2/
`neu–positive cells. The internalization efficiency of the
`
`ELISA of GrB-based fusions on Me180 cells
`0.8
`
`GrB/4D5
`GrB/4D5/26
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`100
`10–3 10–2 10–1
`Conc. (nmol/L)
`
`101
`
`102
`
`Absorbance at 405 nm
`
`GrB/4D5
`GrB/4D5/26
`Control
`
`ELISA of GrB-based fusions on Her2/neu ECD
`1.8
`1.5
`1.2
`0.9
`0.6
`0.3
`0.0
`100
`10–3 10–2 10–1
`Conc. (nmol/L)
`
`101
`
`102
`
`Absorbance at 405 nm
`
`A
`
`Absorbance at 405 nm
`
`ELISA of GrB-based fusions on Her2/neu ECD
`1.8
`1.5
`1.2
`0.9
`0.6
`0.3
`0.0
`100
`10–3 10–2 10–1
`Conc. (nmol/L)
`
`GrB/4D5
`GrB/4D5/26
`Control
`
`101
`
`102
`
`C
`
`PBS
`
`GrB/4D5
`
`GrB/4D5/26
`
`GrB standard
`Pro-GrB/4D5
`GrB/4D5
`Pro-GrB/4D5/26
`GrB/4D5/26
`
`BT474 M1
`
`Me 180
`
`0
`
`10
`
`20
`
`40
`30
`Time (min)
`
`50
`
`60
`
`70
`
`D
`
`GrB/4D5
`
`GrB/4D5/26
`
`0 0.5 1 2 4 8 12 24 36 48 h
`
`0 0.5 1 2 4 8 12 24 36 48 h
`
`Full-length GrB fusion
`
`Free GrB
`β-Actin
`
`0.20
`
`0.15
`
`0.10
`
`0.05
`
`0.00
`
`B
`
`Absorbance at 405 nm
`
`Figure 2. Characterization and comparison of GrB-based fusion proteins. A, Kd of the fusion constructs to Her2/neu ECD, Her2/neu–positive BT474 M1 cells,
`and Her2/neu–negative Me180 cells by ELISA. B, enzymatic activity of GrB moiety of fusion proteins compared with native GrB. C, internalization
`analysis of BT474 M1 cells and Me180 cells after 4 hours of treatment with 25 nmol/L functionalized GrB fusions. Cells were subjected to immunofluorescent
`staining with anti-GrB antibody (FITC-conjugated secondary), with PI nuclear counterstaining. D, Western blot analysis of intracellular behavior of
`25 nmol/L GrB fusion constructs in BT474 M1 cells.
`
`982
`
`Mol Cancer Ther; 12(6) June 2013
`
`Molecular Cancer Therapeutics
`
`Downloaded from
`
`mct.aacrjournals.org
`
`on July 26, 2014. © 2013 American Association for Cancer Research.
`
`IMMUNOGEN 2015, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst March 14, 2013; DOI: 10.1158/1535-7163.MCT-13-0002
`
`Novel Functionalized GrB-Targeting Her2/neu
`
`Table 1. Comparative IC50 values of GrB-based fusion constructs against various types of tumor cell lines
`IC50 (nmol/L)
`
`PI-9
`level

`þþþþþ


`
`Her2/neu
`GrB/4D5/26
`level
`Cell line
`Type
`þþþþ
`29.3
`BT474 M1
`Breast
`þþþþ
`40.5
`Calu3
`Lung
`þþþþ
`90.4
`NCI-N87
`Gastric
`þþþ
`56.8
`MDA MB453
`Breast
`þþ
`93.1
`eB-1
`Breast

`>500.0
`MDA MB435
`Breast


`>500.0
`Me180
`Cervical
`NOTE: þ, indicates the Her2/neu expression level in different cancer cells.
`
`—
`
`—
`
`GrB/4D5
`253.3
`242.4
`629.0
`436.0
`551.3
`>750.0
`>750.0
`
`GrB/26
`905.5
`863.0
`1,106.0
`694.2
`1,134.5
`1,031
`>1,500.0
`
`GrB
`>1,500.0
`>1,500.0
`>1,500.0
`>1,500.0
`>1,500.0
`>1,500.0
`>1,500.0
`
`fusions was further examined by time-dependent West-
`ern blot analysis of the GrB signal (full-length GrB fusion
`þ free GrB; Fig. 2D). Both constructs internalized rapidly
`into BT474 M1 cells within 30 minutes. Compared with
`GrB/4D5, GrB/4D5/26 displayed enhanced and long-
`lasting cell internalization. The intracellular delivery of
`GrB after endocytosis of GrB/4D5 or GrB/4D5/26 also
`was assessed by time-dependent Western blotting (free
`GrB). We observed no GrB delivery by GrB/4D5 up to 48
`hours of treatment, whereas GrB delivery by GrB/4D5/26
`was observed starting at approximately 4 hours of treat-
`ment and presented a tremendously high level of free GrB
`up to 48 hours (Fig. 2D).
`
`In vitro cytotoxic effects of GrB-based fusions
`GrB-based fusions were then tested against a number of
`tumor cell lines. After 72 hours exposure, GrB/4D5/26
`showed specific cytotoxicity to Her2/neu–positive cells,
`with IC50 values of less than 100 nmol/L (Table 1), and
`GrB/4D5 showed cytotoxic effects at somewhat higher
`doses (>200 nmol/L). In addition, GrB/26 showed min-
`imal cytotoxicity at doses more than 600 nmol/L, but no
`significant activity of GrB itself was observed at doses up
`to 1.5 mmol/L. When Her2/neu–positive MDA MB453
`cells were pretreated with Herceptin (5 mmol/L) for 6
`hours and then treated with GrB/4D5/26 for 72 hours, the
`cytotoxicity of GrB/4D5/26 was reduced (Supplementary
`
`Fig. S1), thereby showing a requirement for antigen bind-
`ing of the GrB/4D5/26 construct.
`We further investigated the expression levels of the
`endogenous PI-9 in different tumor cells (Supplementary
`Fig. S2 and Table 1). These studies failed to find an
`association between the response of cells to the cytotox-
`icity of the GrB constructs and the endogenous expression
`of PI-9. This may suggest that factors other than PI-9 may
`account for the observed differences in GrB/4D5/26 cyto-
`toxicity to Her2/neu–expressing target cells.
`
`Cytotoxic effects of GrB/4D5/26 against cells
`resistant to Herceptin or lapatinib
`Acquired resistance to Herceptin or lapatinib can be
`mediated by concomitant upregulation of Her2/neu
`downstream signaling pathways or activation of signaling
`through the ER pathway (30). In this study, we developed
`a model of Herceptin- and lapatinib-resistant variants of
`BT474 M1 cells. Parental BT474 M1 cells were readily
`sensitive to both Herceptin (IC50, 52.5 nmol/L) and lapa-
`tinib (IC50, 34.7 nmol/L; Table 2). Herceptin-resistant cells
`showed resistance to Herceptin [IC50, 10.1 mmol/L; fold
`resistance (FR), 192] but remained sensitive to lapatinib
`(IC50, 32.4 nmol/L). Lapatinib-resistant cells showed
`resistance to high micromolar concentrations of both
`Herceptin (IC50, 74.1 mmol/L; FR, 1411) and lapatinib
`(IC50, 8.2 mmol/L; FR, 237). As shown in Table 2, cells
`
`Table 2. Cytotoxic effects of Her2/neu–targeted therapeutic agents on IC50 values in BT474 M1 cells and
`resistant variants
`
`IC50 (nmol/L) with (FR)a
`
`Agent
`Herceptin
`Lapatinib
`GrB/4D5/26
`
`BT474 M1
`52.5 (1)
`34.7 (1)
`32.9 (1)
`
`BT474 M1
`Herceptin-
`resistant
`10,100.5 (192)
`32.4 (1)
`26.8 (1)
`
`BT474 M1
`lapatinib-
`resistant
`74,100.0 (1,411)
`8,225.0 (237)
`66.1 (2)
`
`BT474 M1 þ
`EGFb
`26,305.0 (501)
`543.0 (16)
`21.7 (1)
`
`BT474 M1 þ
`NRG-1c
`23,033.0 (439)
`547.1 (16)
`18.1 (1)
`
`BT474 M1 þ
`b-estradiold
`74.1 (1)
`33.9 (1)
`31.3 (1)
`
`aFR represents IC50 of agent on BT474 M1–resistant variants/that on BT474 M1 parental cells.
`b,c,dCells were pretreated with b20 ng/mL EGF, c50 ng/mL NRG-1, or d10 ng/mL b-estradiol for 72 hours before drug treatment.
`
`www.aacrjournals.org
`
`Mol Cancer Ther; 12(6) June 2013
`
`983
`
`Downloaded from
`
`mct.aacrjournals.org
`
`on July 26, 2014. © 2013 American Association for Cancer Research.
`
`IMMUNOGEN 2015, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst March 14, 2013; DOI: 10.1158/1535-7163.MCT-13-0002
`
`Cao et al.
`
`resistant to Herceptin showed equivalent sensitivity to the
`
`GrB/4D5/26 construct (IC50, 30 nmol/L for both Her-
`
`ceptin-resistant and parental BT474 M1 cells, respective-
`ly). For lapatinib-resistant cells, the IC50 was marginally
`increased (2-fold) compared with parental cells (66.1 vs.
`32.9 nmol/L).
`We also showed that addition of EGF or neuregulin-1
`(NRG-1) growth factor, but not b-estradiol, to BT474
`M1 parental cells can circumvent the cellular cytotoxic
`responses to Herceptin and lapatinib. Seventy-two hours
`of pretreatment of BT474 M1 cells with 20 ng/mL EGF or
`50 ng/mL NRG-1 resulted in a 400- to 500-fold increase in
`resistance to Herceptin and a 16-fold increase in resistance
`to lapatinib (Table 2). However, treatment of these resis-
`tant cells resulted in no cross-resistance to GrB/4D5/26
`fusions compared with parental BT474 M1 cells.
`A significant observation was that incubation of cells
`with GrB/4D5/26 in the presence of chloroquine did
`not improve cytotoxicity toward these cells (Supple-
`mentary Fig. S3). This finding showed that the fuso-
`
`genic peptide 26 efficiently releases GrB fusion pro-
`teins from intracellular vesicles,
`thereby allowing
`access to cytosolic GrB substrates and induction of
`apoptosis.
`
`Mechanistic studies of GrB/4D5/26 cytotoxicity
`We conducted a panel of experiments to assess the
`potential of GrB-based fusions to initiate the proteolytic
`cascade culminating in apoptosis of BT474 M1 parental,
`Herceptin-, and lapatinib-resistant cells.
`Annexin V/PI staining. Cells were incubated for 24 or
`48 hours with 100 nmol/L GrB/4D5/26 and apoptosis
`was detected via Annexin V/PI staining. GrB/4D5/26
`induced apoptosis in BT474 M1 parental, Herceptin- and
`lapatinib-resistant cells, as indicated by the reduced viable
`population combined with greater populations of early
`apoptosis (Fig. 3A). No apoptosis was induced by 100
`nmol/L GrB/4D5 in any of these cells (Supplementary
`Fig. S4). Her2/neu–negative Me180 cells were not affected
`by either construct.
`
`Figure 3. Effects of GrB-based fusions on apoptotic pathways of BT474 M1 parental, Herceptin-, and lapatinib-resistant cells. A, detection of apoptosis of
`GrB/4D5/26 by Annexin V/PI staining assay. Me180 cells served as a Her2/neu–negative control group. B, Western blot analysis of cleavage and
`activation of caspases-3 and -9 as well as PARP by GrB-based fusion constructs. C, Western blot analysis of apoptosis kinetics and specificity of GrB/4D5/26.
`Cells were treated with GrB/4D5/26 for up to 24 hours with or without 100 mmol/L zVAD-fmk for 24 hours in parental or Herceptin-resistant cells and for up to
`48 hours in lapatinib-resistant cells.
`
`984
`
`Mol Cancer Ther; 12(6) June 2013
`
`Molecular Cancer Therapeutics
`
`Downloaded from
`
`mct.aacrjournals.org
`
`on July 26, 2014. © 2013 American Association for Cancer Research.
`
`IMMUNOGEN 2015, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst March 14, 2013; DOI: 10.1158/1535-7163.MCT-13-0002
`
`Novel Functionalized GrB-Targeting Her2/neu
`
`Activation of caspases. Caspase activation in BT474
`M1 cells was detected by Western blot analysis. Treatment
`with GrB/4D5/26 resulted to the cleavage of caspase-3,
`9, and PARP in all cells, but no activation occurred when
`cells were treated with GrB/4D5 (Fig. 3B). Compared with
`BT474 M1 parental and Herceptin-resistant cells, the acti-
`vations of caspase-9, -3, and PARP were delayed in lapa-
`tinib-resistant cells, which coincided with the observed
`decreased cytotoxic effects.
`We further assessed the kinetics of PARP cleavage
`induced by GrB/4D5/26 on BT474 M1 parental, Her-
`ceptin- and lapatinib-resistant cells, and found that
`cleavage occurred after 2 hours of drug exposure for
`parental and Herceptin-resistant cells but at 24 hours for
`lapatinib-resistant cells (Fig. 3C). In addition, in the
`presence of the pan-caspase inhibitor zVAD-fmk, PARP
`
`cleavage of GrB/4D5/26 was partially inhibited in all
`cells. This finding is in agreement with a mechanism
`relying on GrB activity for caspase-3 cleavage followed
`by PARP cleavage.
`Impact on mitochondrial pathways. We detected cell
`death induced by GrB/4D5/26 via several mitochondrial-
`related pathways. In BT474 M1 parental, Herceptin- and
`lapatinib-resistant cells, GrB/4D5/26 treatment activated
`BID and downregulated the antiapoptotic Bcl-2 protein
`(Fig. 4A), and it triggered the release of cytochrome c from
`the mitochondria into the cytosol (Fig. 4B). Bax was
`normally present in both the cytosol and mitochondria
`of untreated cells. However, when the cells were treated
`with GrB/4D5/26, Bax was decreased in cytosol and
`increased in mitochondria (Fig. 4B). As previously
`described, treatment for 24 hours with GrB/4D5/26 was
`
`Figure 4. Effects of GrB fusion
`constructs on mitochondrial
`pathway in BT474 M1 parental,
`Herceptin-, and lapatinib-resistant
`cells. A, effects of GrB-based fusion
`proteins on the upstream
`components Bcl-2 and BID in
`mitochondrial pathway. B, effects of
`the fusion constructs on cytochrome
`c release and Bax translocation.
`
`www.aacrjournals.org
`
`Mol Cancer Ther; 12(6) June 2013
`
`985
`
`Downloaded from
`
`mct.aacrjournals.org
`
`on July 26, 2014. © 2013 American Association for Cancer Research.
`
`IMMUNOGEN 2015, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst March 14, 2013; DOI: 10.1158/1535-7163.MCT-13-0002
`
`Cao et al.
`
`Figure 5. Western blot analyses of
`the effects of GrB/4D5 and GrB/
`4D5/26 in BT474 M1 parental,
`Herceptin-, and lapatinib-resistant
`cells on Her2/neu and ER signaling
`pathways. Cells were treated with
`100 nmol/L functionalized GrB
`constructs for 24 or 48 hours, and
`total cell lysates were quantified
`and further measured by Western
`blot analysis of pHer2/neu, pAkt,
`pmTOR, pERK, ER, PR, and PI-9
`level.
`
`shown to activate the mitochondrial pathway in both
`BT474 M1 parental and Herceptin-resistant cells, but this
`activation was delayed in lapatinib-resistant cells.
`
`Effects of GrB fusions on Her- and ER-associated
`signaling pathways
`We next examined the mechanistic effects of the con-
`structs on Her- and ER-related signaling events in BT474
`M1 parental cells and the resistant variants. As shown in
`Supplementary Fig. S5, cells resistant to Herceptin had
`enhanced Her family receptor activity but reduced levels
`of PR and PI-9. In contrast, in lapatinib-resistant cells there
`was total downregulation of Her family receptor activity
`but higher levels of ER, PR, and PI-9.
`Cells treated with GrB/4D5 or GrB/4D5/26 showed the
`effects on these signaling pathways, corresponding to the
`cytotoxic results we observed (Fig. 5). Treatment with
`GrB/4D5/26 remarkedly inhibited phosphorylation of
`Her2/neu and its downstream molecules Akt, mTOR,
`and ERK, which are critical events in Her2/neu signaling
`cascade. In contrast, GrB/4D5 showed a comparatively
`reduced effect on these pathways. We observed a reduced
`ER level among all cells. Evidence from other researchers
`
`has shown that upregulation of the ER pathway in ER-
`and Her2/neu–positive cell lines with lapatinib creates
`an escape/survival pathway (30, 31), but GrB/4D5/26
`seem to be able to inactivate all the signaling pathways
`in these cells. We also observed the delaying signaling
`effects of GrB/4D5/26 on lapatinib-resistant cells com-
`pared with parental or Herceptin-resistant cells, which
`was in agreement with the apoptotic cell death results
`observed for the lapatinib-resistant cells. Notably, there
`was an increased mRNA and protein level of PI-9 in this
`resistant line but not in the parental or Herceptin-resis-
`tant cells (Supplementary Figs. S5 and S6). Taken
`together, these results suggest that activation of the ER
`pathway upregulates the expression of PI-9 and this
`results in a slight inhibition of GrB/4D5/26 activity and
`a delay in apoptotic cell death compared with parental
`cells.
`Our investigation suggests that the GrB/4D5/26 fusion
`is more cytotoxic than GrB/4D5 construct to Her2/
`neu–positive cells, even those that have acquired resis-
`tance to the traditional Her2/neu therapeutic agents Her-
`ceptin and lapatinib. The cytotoxicity results coincide
`with the observed effects on signal transduction and
`
`986
`
`Mol Cancer Ther; 12(6) June 2013
`
`Molecular Cancer Therapeutics
`
`Downloaded from
`
`mct.aacrjournals.org
`
`on July 26, 2014. © 2013 American Association for Cancer Research.
`
`IMMUNOGEN 2015, pg. 8
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst March 14, 2013; DOI: 10.1158/1535-7163.MCT-13-0002
`
`Table 3. Cytotoxicity of chemical agents and
`GrB-based fusions on MDR-1–expressing cells
`
`IC50 (nmol/L)
`
`BT474
`M1
`5.2
`1.3
`311.8
`34.1
`
`BT474 M1
`MDR-1
`1,047.3
`105.1
`318.9
`35.5
`
`FRa
`209
`89
`1
`1
`
`Taxol
`Vinblastin
`GrB/4D5
`GrB/4D5/26
`
`aFR represents IC50 of agent on BT474 M1 MDR-1 cells/that
`on BT474 M1 parental cells.
`
`monitoring these pathways may be useful as a monitor of
`drug efficacy.
`
`Effects of GrB/4D5/26 on the MDR-1–expressing
`cells
`Multidrug resistance (MDR) is a phenomenon, which
`results from various reasons. The most-characterized
`cause of MDR is the overexpression of a 170-kDa mem-
`brane glycoprotein known as P-glycoprotein (Pgp). To
`verify the effects of GrB-based fusi

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