throbber
Published OnlineFirst November 24, 2009; DOI: 10.1158/0008-5472.CAN-09-2693
`Published OnlineFirst November 24, 2009; DOI: 10.1158/0008-5472.CAN-09-2693
`
`Construction and Characterization of Novel, Recombinant
` Oncogene Product:
`Her2/neu
`Immunotoxins Targeting the
` Studies
`In vivo
` and
`vitro

`Yu Cao, James D. Marks, John W. Marks, et al.
`Cancer Res  
`
`2009;69:8987-8995. Published OnlineFirst November 24, 2009.
`
`In
`




`


`
`Updated version

`Supplementary
`Material

`
`Access the most recent version of this article at:
`
`10.1158/0008-5472.CAN-09-2693
`doi:

`Access the most recent supplemental material at:
`http://cancerres.aacrjournals.org/content/suppl/2009/11/24/0008-5472.CAN-09-2693.DC1.ht
` ml

`
`Cited Articles

`Citing articles

`
`This article cites by 47 articles, 16 of which you can access for free at:
`http://cancerres.aacrjournals.org/content/69/23/8987.full.html#ref-list-1

`This article has been cited by 6 HighWire-hosted articles. Access the articles at:
`
`http://cancerres.aacrjournals.org/content/69/23/8987.full.html#related-urls

`
`
`
`E-mail alerts

`Reprints and
`Subscriptions

`Permissions

`
`Sign up to receive free email-alerts
`
` related to this article or journal.
`
`To order reprints of this article or to subscribe to the journal, contact the AACR Publications
`.
`pubs@aacr.org
`Department at

`To request permission to re-use all or part of this article, contact the AACR Publications
`.
`permissions@aacr.org
`Department at

`
`Downloaded from
`Downloaded from
`
`on October 7, 2014. © 2009 American Association for Cancercancerres.aacrjournals.org
`
`
`on October 7, 2014. © 2009 American Association for Cancercancerres.aacrjournals.org
`Research.
`Research.
`
`IMMUNOGEN2111, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst November 24, 2009; DOI: 10.1158/0008-5472.CAN-09-2693
`
`Experimental Therapeutics, Molecular Targets, and Chemical Biology
`
`Construction and Characterization of Novel, Recombinant
`Immunotoxins Targeting the Her2/neu Oncogene Product:
`In vitro and In vivo Studies
`
`Yu Cao,1 James D. Marks,2 John W. Marks,1 Lawrence H. Cheung,1 Sehoon Kim,1
`and Michael G. Rosenblum1
`
`1Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics, M.D. Anderson Cancer Center,
`Houston, Texas and 2Department of Anesthesia, University of California, San Francisco, San Francisco, California
`
`Abstract
`The goal of this study was to characterize a series of anti-
`Her2/neu immunotoxin constructs to identify how different
`antibodies and linker choices affect the specificity and cyto-
`toxicity of these proteins. We constructed a series of immuno-
`toxins containing either the human single-chain antibody
`(scFv) C6.5 or the murine scFv e23 fused to the highly toxic
`recombinant gelonin (rGel) molecule. Based on the flexible
`GGGGS linker (L), the fusion construct C6.5-L-rGel was com-
`pared with e23-L-rGel to evaluate the specific cytotoxic effects
`against Her2/neu-positive and Her2/neu-negative tumor cells.
`Both constructs retained the specificity of the original anti-
`body as well as the biological activity of rGel toxin. The two
`constructs displayed similar cytotoxicity against different car-
`cinoma cells. We additionally introduced the modified linkers
`TRHRQPRGWEQL (Fpe) and AGNRVRRSVG (Fdt), which
`contained furin cleavage sites, to determine the effect of these
`design changes on stability and cell killing efficiency. The in-
`troduction of furin cleavage linkers (Fpe or Fdt) into the mo-
`lecules resulted in dissimilar sensitivity to protease cleavage
`compared with the constructs containing the L linker, but
`very similar intracellular rGel release, cytotoxic kinetics,
`and induction of autophagic cell death in vitro. Xenograft
`studies with SKOV3 ovarian tumors were done using various
`C6.5/rGel constructs. C6.5-L-rGel was more efficient in tumor
`inhibition than constructs containing furin linkers, attribut-
`ing to a higher stability in vivo of the L version. Therefore,
`our studies suggest that human C6.5-L-rGel may be an effec-
`tive novel clinical agent for therapy of patients with Her2/neu-
`overexpressing malignancies. [Cancer Res 2009;69(23):8987–95]
`
`Introduction
`The Her2/neu proto-oncogene encodes a 185-kDa transmem-
`brane glycoprotein kinase with extensive homology to the epider-
`mal growth factor receptor (HER1; refs. 1–3). Amplification of
`the gene and overexpression of the Her2/neu protein product in
`tumor cells have been well described in numerous human cancers,
`
`Note: Supplementary data for this article are available at Cancer Research Online
`(http://cancerres.aacrjournals.org/).
`Current address for S. Kim: Merck & Co., Inc., 21 Lafayette, Lebanon, NH 03766.
`Requests for reprints: Michael G. Rosenblum, M.D. Anderson Cancer Center, Box
`0044, 1515 Holcombe Boulevard, Houston, TX 77030. Phone: 713-792-3554; Fax:
`713-794-4261; E-mail: mrosenbl@mdanderson.org.
`©2009 American Association for Cancer Research.
`doi:10.1158/0008-5472.CAN-09-2693
`
`including mammary and ovarian carcinomas and gastric and lung
`tumors (4–6). Because Her2/neu plays a central role in malignant
`transformation and growth, it provides an attractive target for
`focused therapeutic approaches.
`A number of approved immunotherapeutic agents are directed
`at tumors that express high levels of Her2/neu, such as the mono-
`clonal antibody trastuzumab (Herceptin), and small-molecule inhi-
`bitors such as gefitinib (Iressa) have shown promising results, but
`the development of resistance to treatment remains a well-known
`problem (7, 8). To enhance its clinical potential, cell-surface Her2/
`neu has been targeted using antibody-drug conjugates (9) or im-
`munotoxins, composed of plant or bacterial toxins linked with a
`targeting molecule composed of monoclonal antibodies or anti-
`body fragments (10, 11). Previously, a recombinant, murine anti-
`Her2/neu single-chain antibody (scFv), designated e23, has been
`fused to catalytic toxins such as Pseudomonas exotoxin A and diph-
`theria toxin (DT) to specifically target Her2/neu-expressing cells
`(12, 13). A major drawback of such proteins is their potential for
`immune response after repeated administration. Further complica-
`tions could result from nonspecific binding of foreign proteins to
`vascular endothelial cells, leading to vascular leak syndrome and,
`ultimately, interstitial edema and organ failure (14, 15).
`The development of immunotoxins containing human or hu-
`manized components may circumvent these problems. Such im-
`munotoxins may display reduced immunogenicity although
`antibodies to the toxin components may still limit prolonged ther-
`apy (16). We previously reported in vitro characterization and
`in vivo antitumor efficacy studies of an immunotoxin composed
`of the human chimeric anti-Her2/neu antibody (BACH-250) chem-
`ically conjugated to recombinant gelonin (rGel; ref. 17). rGel is a
`29-kDa ribosome-inactivating plant toxin with a potency and
`mechanism of action similar to that of ricin toxin A-chain, but with
`improved stability and reduced toxicity (18, 19). The BACH-250/
`rGel conjugate showed potent and specific cytotoxicity against
`Her2/neu-overexpressing human tumor cells in culture and against
`SKOV3 tumor xenografts. However, the treatment of solid tumors
`presents a potential problem because full-length antibodies must
`diffuse into the tumor against a hydrostatic pressure gradient and
`into disordered vasculature (20, 21).
`Marks and colleagues previously described an anti-Her2/neu
`scFv, designated C6.5, which was selected from a human scFv
`phage display library and affinity-matured in vitro (22). Using scFv
`C6.5, McCall and colleagues constructed and characterized a bis-
`pecific scFv composed of C6.5 and anti-CD16 scFv, displaying a
`high level of in vitro tumor cell cytotoxicity and in vivo tumor tar-
`geting (23). Studies by Park and colleagues generated immunolipo-
`somes containing doxorubicin, which were targeted to tumor cells
`
`www.aacrjournals.org
`
`8987
`
`Cancer Res 2009; 69: (23). December 1, 2009
`
`
`
`cancerres.aacrjournals.org Downloaded from
`
`on October 7, 2014. © 2009 American Association for Cancer
`Research.
`
`IMMUNOGEN2111, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst November 24, 2009; DOI: 10.1158/0008-5472.CAN-09-2693
`
`Cancer Research
`
`using antibody C6.5. These constructs showed selective enhance-
`ment of the therapeutic index of doxorubicin chemotherapy (24).
`Most recently, Adams and colleagues successfully used a C6.5 dia-
`body construct as a radioimmunotherapeutic agent containing
`(211At) for the treatment of Her2/neu-positive solid tumors in xe-
`nograft models, showing that scFv C6.5 could be used effectively in
`vehicles for targeted radioimmunotherapy by using powerful,
`short-lived α-emitting radioisotopes (25).
`In the present study, we describe the construction and charac-
`terization of several rGel-based chimeric toxins composed of the
`scFv e23 or C6.5 and using various linker configurations to exam-
`ine how different antibodies and linker choices affect the in vitro
`and in vivo efficacy of fusion constructs.
`
`Materials and Methods
`Plasmid construction. The gene encoding murine scFv e23 was ob-
`tained from Oncologix, Inc., and human scFv C6.5 was kindly supplied by
`Prof. James D. Marks (University of California, San Francisco, San Francisco,
`CA). Illustrations of the immunotoxin constructs are shown in Fig. 1A.
`Recombinant immunotoxins containing rGel and either e23 or C6.5 and
`the flexible linker (GGGGS, designated “L”) were constructed by overlapping
`PCR. These proteins were designated e23-L-rGel and C6.5-L-rGel, respec-
`tively. In addition, we generated two different linkers (TRHRQPRGWEQL,
`designated “Fpe,” and AGNRVRRSVG, designated “Fdt”) containing furin
`cleavage sites from Pseudomonas exotoxin A and DT, incorporated between
`the C6.5 and rGel components. The intent was that furin cleavage would
`allow more facile release of the toxin from the complex once internalized
`by the endosomal compartment. The resulting proteins were designated
`C6.5-Fpe-rGel and C6.5-Fdt-rGel, respectively. All the fusion genes were
`cloned into a T7 promoter–based E. coli expression vector, pET-32a(+).
`Protein expression in E. coli. To express the recombinant fusion pro-
`teins, bacterial cultures were incubated at 37°C in LB growth medium with
`strong antibiotic selection (400 μg/mL ampicillin, 70 μg/mL chloramphen-
`icol, and 15 μg/mL kanamycin) and grown to log phase (A600 nm = 0.8). The
`target protein expression was induced at 18°C with 0.1 mmol/L isopropyl-L-
`thio-β-D-galactopyranoside for 16 h. Induced bacterial cultures were then
`centrifuged and stored frozen at −80°C.
`Isolation and purification of immunotoxins. Frozen bacterial pellets
`containing different immunotoxins were purified as follows: The pellets
`were allowed to thaw with the addition of 50 mmol/L Na-phosphate (pH
`7.6), 300 mmol/L NaCl, 5 mmol/L imidazole. The bacterial suspension was
`further lysed by passing the material through a microfludizer (Microflui-
`dics). The solution was then clarified by centrifugation (40,000 rpm), and
`the supernatants were loaded onto columns (2.5-cm internal diameter × 13
`cm) containing cobalt-charged IMAC resins. After washing with 50 mmol/L
`Na-phosphate (pH 7.6), 300 mmol/L NaCl, 15 mmol/L imidazole, the bound
`protein was eluted with 50 mmol/L Na-phosphate (pH 7.6), 300 mmol/L
`NaCl, 300 mmol/L imidazole. Fractions containing immunotoxin were
`dialyzed in 20 mmol/L Tris-HCl (pH 7.4) and 150 mmol/L NaCl, followed
`by digestion with recombinant enterokinase. The purified immunotoxins
`were dialyzed in PBS, filter sterilized, and stored at 4°C.
`Binding affinity and internalization analyses. The binding affinity
`and specificity of rGel-based immunotoxins containing either scFv e23
`or C6.5 were evaluated by ELISA on Her2/neu-positive SKOV3 cells and
`Her2/neu-negative MCF7 cells. Rabbit anti-rGel antibody and horseradish
`peroxidase–conjugated goat anti-rabbit IgG was used as a tracer in this
`assay as described previously (26).
`Immunofluorescence-based internalization studies were also done on
`SKOV3 and MCF7 cells (27). Cells treated with immunotoxins or rGel
`were subjected to immunofluorescent staining with anti-rGel antibody
`(FITC-conjugated secondary antibody). Nuclei were counterstained with
`propidium iodine. Visualization of immunofluorescence was done with a
`confocal laser scanning microscope (Zeiss LSM510, Carl Zeiss).
`Reticulocyte lysate in vitro translation assay. The rGel-induced
`inhibition of [3H]leucine incorporation into protein in a cell-free protein
`
`Figure 1. Preparation of e23/rGel and C6.5/rGel series immunotoxins. A,
`schematic diagram of immunotoxin constructs containing scFv (e23 or C6.5),
`peptide linker (L, Fpe, or Fdt), and rGel toxin. B, SDS-PAGE analysis of purified
`immunotoxins.
`
`synthesizing system after the administration of various doses of immuno-
`toxins was done as described previously (28).
`Furin cleavage assay. Various C6.5/rGel fusions containing different
`linkers were treated with recombinant human furin (NEB) at various pH.
`For pH 5.4, 50 mmol/L sodium acetate buffer was used, and for pH 7.2, 10
`mmol/L HEPES buffer was used. A dose of 2 units of purified furin was
`applied to 25 μg of fusion protein in each reaction. After incubation for
`16 h at room temperature, the proteins were analyzed by Western blot with
`anti-rGel antibody. The rate of cleavage by furin was calculated using
`AlphaEaseFC software (version 4.0.1) from the following formula: cleavage
`rate = value of gray scale of cleaved protein / value of gray scale of total
`protein.
`In vitro cytotoxicity assays. Log-phase cells were seeded (∼5 × 103 per
`well) in 96-well plates and allowed to attach overnight. Cells were further
`incubated with various concentrations of immunotoxins, rGel, or medium
`at 37°C for 72 h, and cell viability was determined by using the crystal violet
`staining method followed by solubilization of the dye in Sorensor's buffer as
`described previously (26).
`Intracellular release of rGel. The intracellular rGel release from C6.5/
`rGel fusion constructs was analyzed in SKOV3 cells. After incubation for
`various times, the cells were treated with acidic glycine buffer [500
`mmol/L NaCl and 0.1 mol/L glycine (pH 2.5)] to strip cell-surface–bound
`proteins and lysed in lysis buffer [10 mmol/L Tris-HCl (pH 8.0), 60 mmol/L
`KCl, 1 mmol/L EDTA, 1 mmol/L DTT, 0.2% NP40]. Cytosolic fractions were
`further quantified and analyzed by Western blot using anti-rGel antibody.
`Western blot analysis of apoptosis and autophagy. SKOV3 cells
`treated with 200 nmol/L immunotoxins were pelleted and lysed. Proteins
`
`Cancer Res 2009; 69: (23). December 1, 2009
`
`8988
`
`www.aacrjournals.org
`
`Downloaded from
`
`cancerres.aacrjournals.org
`
`on October 7, 2014. © 2009 American Association for Cancer
`Research.
`
`IMMUNOGEN2111, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst November 24, 2009; DOI: 10.1158/0008-5472.CAN-09-2693
`
`Novel Immunotoxins Targeting Her2/neu
`
`from each cell lysates were analyzed by Western blot with antibodies
`that recognized poly(ADP-ribose) polymerase (PARP), β-actin, high mo-
`bility group box 1 (HMGB1; Santa Cruz Biotechnology), and microtu-
`bule-associated protein 1 light chain 3 (MAP LC3; Novus Biological).
`For HMGB1 release assay, the medium was harvested, concentrated,
`and further analyzed by Western blot.
`In vivo efficacy studies. BALB/c nude mice were injected s.c. with
`SKOV3 cells (5 × 106 per mouse). Once tumors were measurable (∼40–50
`mm3), animals were treated (i.v. via tail vein) with PBS or rGel as control or
`with different C6.5/rGel fusion proteins, every other day for 10 d. Animals
`were monitored and tumors were measured for an additional 40 d.
`Immunofluorescence analysis. Twenty-four hours after i.v. injection of
`C6.5-L-rGel or rGel, the mice were sacrificed and tumor samples were col-
`lected and frozen immediately for sectioning. The sample slides were fixed
`in 3.7% paraformaldehyde, permeabilized with PBS containing 0.2% Triton
`X-100, and blocked in 5% nonfat milk in PBS. After incubation with anti-
`rGel antibody, the samples were subjected to immunofluorescent staining
`with FITC-conjugated secondary antibody and nuclear counterstaining
`with propidium iodine. The slides were mounted and delivered for the im-
`munofluorescence observation under a Nikon Eclipse TS-100 fluorescence
`microscope (Nikon).
`
`the native rGel NH2 terminus. VH/VL orientations determined the
`best binding activity of VL-VH for e23 and VH-VL for C6.5 (data not
`shown). The C6.5/rGel construct was further engineered by incor-
`porating two different enzymatically sensitive furin cleavage lin-
`kers between the scFv and rGel toxin components. The two furin
`sensitive sequences are designated Fpe (TRHRQPRGWEQL) and
`Fdt (AGNRVRRSVG; Fig. 1A). Several biochemical studies have
`shown that the serine protease furin efficiently cleaves proteins
`containing these recognition sequences (29, 30).
`Following purification, all the rGel-based immunotoxins migrat-
`ed on SDS-PAGE at the expected molecular weight of 55 kDa
`(Fig. 1B). However, with the introduction of sensitive furin linker,
`C6.5-Fdt-rGel, but not C6.5-Fpe-rGel, displayed cleavage bands to
`some extent after rEK digestion. The cleavage was found to be oc-
`curring precisely at the predicted furin cleavage site producing the
`27- to 28-kDa fragments of scFv and rGel. Further analysis indicat-
`ed that the yields for each protein (per liter of bacterial culture)
`were 1.55 mg for e23-L-rGel, 1.05 mg for C6.5-L-rGel, 1.08 mg for
`C6.5-Fpe-rGel, and 0.70 mg for C6.5-Fdt-rGel.
`
`Results
`
`Construction, expression, and purification of rGel-based fu-
`sions. The initial rGel-based immunotoxins consisted of a flexible
`L linker (GGGGS) tethering the COOH terminus of e23 or C6.5 to
`
`Characterization of e23-L-rGel and C6.5-L-rGel
`Immunotoxins
`Binding activity. To ensure that immunotoxins retained anti-
`gen binding ability, the fusion proteins were compared in an
`ELISA-based binding assay (Fig. 2A) using Her2/neu-positive
`
`Figure 2. Characterization and comparison of e23-L-rGel and C6.5-L-rGel immunotoxins. A, evaluation of the binding activity of the fusion constructs to SKOV3
`and MCF7 cells by whole-cell ELISA. B, the enzymatic (N-glycosidase) activity of the rGel component of the fusion was assessed using rabbit reticulocyte lysate assay.
`C, internalization of e23-L-rGel and C6.5-L-rGel into SKOV3 and MCF7 cells. Cells were subjected to immunofluorescent staining with anti-rGel antibody
`(FITC-conjugated secondary) with propidium iodine nuclear counterstaining.
`
`www.aacrjournals.org
`
`8989
`
`Cancer Res 2009; 69: (23). December 1, 2009
`
`
`
`cancerres.aacrjournals.org Downloaded from
`
`on October 7, 2014. © 2009 American Association for Cancer
`Research.
`
`IMMUNOGEN2111, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst November 24, 2009; DOI: 10.1158/0008-5472.CAN-09-2693
`
`Cancer Research
`
`SKOV3 and Her2/neu-negative MCF7 cells. The equilibrium disso-
`ciation constant, Kd, was further calculated (GraphPad Prism,
`v4.03). The affinity of e23-L-rGel (Kd = 8.5 nmol/L) for SKOV3 cells
`was similar to that of C6.5-L-rGel (Kd = 12.6 nmol/L). The Kd values
`were consistent with those previously measured in an in vitro live
`cell assay using scFv itself (22). In addition, both immunotoxins
`showed significant specificity based on the background of binding
`to MCF7 cells. ELISA assay suggested that the human scFv C6.5 dis-
`played similar binding specificity compared with the murine e23.
`Cell-free protein synthesis inhibitory activity. The biological
`activity of toxins can be severely compromised when incorporated
`into fusion constructs. To examine the N-glycosidic activity of the
`rGel component of the immunotoxins, these materials were added
`to an in vitro protein translation assay using [3H]leucine incorpo-
`ration by isolated rabbit reticulocytes. Inhibition curves for the fu-
`sion constructs e23-L-rGel and C6.5-L-rGel and native rGel were
`compared (Fig. 2B), and IC50 values for the three molecules were
`found to be virtually identical (15.41, 15.52, and 10.6 pmol/L, re-
`spectively), suggesting that no loss of toxin activity occurred in
`the fusion molecules.
`Cellular uptake of immunotoxins. We next examined whether
`the e23-L-rGel and C6.5-L-rGel fusions could specifically internal-
`ize into target cells. Immunofluorescence staining was done on
`SKOV3 and MCF7 cells after exposure to the constructs. As shown
`in Fig. 2C, the rGel moiety of both fusions was observed primarily
`in the cytosol of SKOV3 after treatment, but not in MCF7 cells,
`showing that both constructs were comparable in efficient cell
`binding and rapid internalization and delivery of rGel toxin to
`the cytoplasm after exposure to Her2/neu-positive cells.
`In vitro cytotoxicity. The e23-L-rGel and C6.5-L-rGel constructs
`and rGel were tested against a number of different tumor cell lines
`(Table 1). The SKBR3 cells with the highest level of Her2/neu ex-
`pression were killed most efficiently by both antibody fusion con-
`structs, with IC50 values of 6.0 and 9.1 nmol/L for e23-L-rGel and
`C6.5-L-rGel, respectively. IC50 values for rGel toxin were ∼200-fold
`higher (1,671 nmol/L). For the other Her2/neu-positive cells, both
`immunotoxins also showed similar IC50 values, showing that the
`two fusion proteins possess very similar cell killing activity and
`specificity. Furthermore, MCF7 and 4T1 cells, which express rela-
`tively low levels of Her2/neu, showed little to no specific cytotoxi-
`city of the fusion constructs compared with rGel itself, clearly
`showing that the presence of higher levels of cell-surface Her2/
`neu is required for specific cytotoxicity of the constructs.
`
`In vitro cleavage of C6.5/rGel fusions by furin. From the
`in vitro study, it was evident that no significant differences were
`observed between murine e23– and human C6.5–based fusion con-
`structs. Therefore, we focused on C6.5/rGel for further studies by
`incorporation of proteolytically cleavable linkers (Fpe and Fdt) to
`examine whether this change would improve killing efficiency. To
`investigate the susceptibility of various chimeric toxins to proteo-
`lytic cleavage, purified fusions were subjected to proteolysis with
`recombinant furin (Fig. 3A). At pH 7.2, cleavage of Fpe (18.5% of
`total) and Fdt (100%) was observed. At pH 5.4, Fpe was cleaved less
`efficiently (4.5% of total), but Fdt still displayed high cleavage effi-
`ciency (100%). In contrast, fusion with L linker was found to be
`highly stable and could not be cleaved at either pH. As indicated,
`the Fdt linker was the most sensitive to cleavage among all the con-
`structs. In contrast, cleavage of the molecule containing the Fpe
`linker was highly dependent on pH. The L linker was found to be
`comparatively resistant to protease action without regard to the pH.
`Kinetics of cytotoxicity by C6.5/rGel fusions. To investigate
`the kinetics of cytotoxicity by different C6.5/rGel fusions, their cell
`killing activities were assessed against SKBR3, SKOV3, Calu3, and
`MDA MB435S cells at various time points (Supplementary
`Table S1). Interestingly, the cell lines showed no differences in
`overall sensitivity to the fusion constructs with furin cleavage lin-
`kers compared to those with flexible L linker. All the fusions
`showed potent cytotoxicity after 48 hours and exerted highly po-
`tent cell killing at 72 hours. This suggests that the cleavage efficien-
`cy of different linkers for these chimeric toxins was not a major
`determinant of the overall cytotoxic effects observed with different
`linkers. Surprisingly, the cytotoxic kinetics of the constructs there-
`fore seemed to be independent of the sensitivity of the constructs
`to proteolytic cleavage.
`Intracellular release of rGel from various constructs. The in-
`tracellular release of rGel after endocytosis of various C6.5/rGel fu-
`sion constructs was assessed by Western blot with an anti-rGel
`antibody (Fig. 3B). During the treatment of SKOV3 cells, rGel re-
`lease was found to be maximal at 2 hours after treatment with
`C6.5-L-rGel and 4 hours after exposure to C6.5-Fpe-rGel. For
`C6.5-Fdt-rGel, the rGel component was released within 1 to 2
`hours and degraded simultaneously, corresponding to the status
`of full-length protein. The decreasing intracellular level of full-
`length C6.5-Fdt-rGel could be ascribed to rapid instability of the
`construct after internalization. Although the maximal rGel release
`of different fusions was achieved at different time points, the
`
`Table 1. Comparative IC50 values of the e23-L-rGel and C6.5-L-rGel fusion constructs against various types of tumor cell lines
`
`Cell line
`
`Origin
`
`Her2/neu level
`
`IC50 (nmol/L)
`
`Targeting index*
`
`e23-L-rGel
`
`C6.5-L-rGel
`
`rGel
`
`e23-L-rGel
`
`C6.5-L-rGel
`
`SKBR3
`NCI-N87
`Calu3
`SKOV3
`BT474
`MDA MB435S
`MCF7
`4T1
`
`Human, breast
`Human, gastric
`Human, lung
`Human, ovarian
`Human, breast
`Human, breast
`Human, breast
`Mouse, breast
`
`High
`High
`High
`High
`High
`Medium
`Low
`No
`
`6.0
`59.2
`41.1
`16.3
`27.1
`24.6
`266.3
`>1,000
`
`9.1
`45.0
`31.3
`18.0
`25.2
`28.8
`200.9
`>1,000
`
`1,671.0
`1,334.0
`879.7
`378.9
`325.2
`359.0
`260.4
`>1,000
`
`279
`23
`21
`23
`12
`15
`1
`1
`
`184
`30
`28
`21
`13
`12
`1
`1
`
`*Targeting index represents IC50 of rGel/ IC50 of immunotoxin.
`
`Cancer Res 2009; 69: (23). December 1, 2009
`
`8990
`
`www.aacrjournals.org
`
`Downloaded from
`
`cancerres.aacrjournals.org
`
`on October 7, 2014. © 2009 American Association for Cancer
`Research.
`
`IMMUNOGEN2111, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst November 24, 2009; DOI: 10.1158/0008-5472.CAN-09-2693
`
`Novel Immunotoxins Targeting Her2/neu
`
`Figure 3. Functional analysis of C6.5/rGel series immunotoxins in vitro. A, Western blot analysis of furin cleavage of purified C6.5/rGel fusion constructs. B, Western
`blot analysis of intracellular rGel release of C6.5/rGel fusions in SKOV3 cells. C, functional stability analysis of the fusions by whole-cell ELISA and cytotoxicity on
`SKOV3 cells. The proteins were incubated in human plasma at 37°C for up to 72 h before analysis.
`
`absolute amounts of delivered rGel found in the cytosol were vir-
`tually identical. Therefore, these data confirm the observation
`that introduction of an unstable furin cleavage linker does not
`improve the intracellular rGel release of the constructs.
`Functional stability analysis of C6.5/rGel fusions. The linkers
`between C6.5 and rGel showed a differential sensitivity to protease
`action, which may result in different clearance and metabolic ki-
`netics in vivo (31, 32). To estimate the stability of various C6.5/rGel
`fusions, we incubated the purified proteins at 37°C for varying
`times in the presence of human plasma before testing cellular
`Her2/neu binding to SKOV3 cells (Fig. 3C). Our results showed that
`in the presence of human plasma, the C6.5-Fdt-rGel construct dis-
`played a reduction in binding activity within 6 hours of incubation
`and a 20% loss of binding activity after 72 hours of incubation. In
`contrast, the C6.5-L-rGel and C6.5-Fpe-rGel fusion constructs
`showed only 9% and 12% reductions, respectively, after 72 hours
`of incubation.
`In addition, the immunotoxins were evaluated for cytotoxic ac-
`tivity following incubation in human plasma for 0, 24, 48, and 72
`hours (Fig. 3C). For the C6.5-Fdt-rGel construct, the cell killing ac-
`tivity was reduced more than 2-fold after 48 hours, as indicated by
`increasing IC50 values of 20 versus 48 nmol/L. However, this was
`not the case for C6.5-L-rGel and C6.5-Fpe-rGel, which retained
`most of its cytotoxic activity even after 48 hours and displayed a
`little influence on IC50 after 72 hours of incubation in plasma (16
`versus 22 nmol/L and 17 versus 25 nmol/L for each construct).
`This functional stability analysis indicated that compared with
`
`the L and Fpe linkers, the Fdt linker was much more unstable in
`human plasma, and this may reduce the in vivo potency of poten-
`tial therapeutic applications using constructs containing this linker
`design.
`Mechanisms of cytotoxic effects. The cytotoxic effects
`mediated by C6.5/rGel fusions were analyzed to evaluate whether
`the cytotoxic mechanisms of the constructs observed included
`elements of apoptosis, necrosis, or autophagy in SKOV3 cells. As
`shown in Fig. 4A, C6.5/rGel fusions did not show activation of
`caspase-dependent apoptosis in SKOV3 cells and showed no cleav-
`age of caspase substrate PARP. The terminal deoxyribonucleotidyl
`transferase–mediated dUTP nick end labeling (TUNEL) results
`(Supplementary Fig. S1) confirmed that the cytotoxic effects of
`the rGel-based fusions were not mediated by apoptosis and DNA
`fragmentation.
`To assess whether the necrotic cell death was induced, we ex-
`amined lactate dehydrogenase (LDH) release, which is a marker of
`abrupt membrane lysis (33). In this case, treatment of SKOV3 cells
`with Triton X-100 serves as a positive control causing LDH release
`(Supplementary Fig. S2). In contrast, treatment with the fusion
`constructs failed to show LDH release, indicating that the observed
`cytotoxicity did not seem to be a result of necrosis.
`We next asked if the immunotoxins activate autophagic sig-
`naling in SKOV3 cells. MAP LC3-I, known to be usually pres-
`ent in the cytosol,
`is palmitoylated during autophagy to form
`membrane-bound LC3-II and is associated with autophagosomes
`(34). As shown in Fig. 4B, the ratio of LC3-II formation to the
`
`www.aacrjournals.org
`
`8991
`
`Cancer Res 2009; 69: (23). December 1, 2009
`
`Downloaded from
`
`cancerres.aacrjournals.org
`
`on October 7, 2014. © 2009 American Association for Cancer
`Research.
`
`IMMUNOGEN2111, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst November 24, 2009; DOI: 10.1158/0008-5472.CAN-09-2693
`
`Cancer Research
`
`β-actin control was increased after treatment with the fusion con-
`structs, showing that autophagic flux was induced by C6.5/rGel
`fusions in SKOV3 cells. In addition, autophagy induction by C6.5/
`rGel fusions was further validated by the selective release of
`HMGB1 (Fig. 4C). Tumor cells that are dying by autophagy selec-
`tively release the nuclear HMGB1 protein without displaying char-
`acteristics of necrosis (35). These data indicated that the observed
`cytotoxic effects of C6.5/rGel fusions on SKOV3 cells seemed to be
`mediated not through an apoptotic or necrotic mechanism but by
`the efficient induction of autophagic cell death.
`Antitumor activity of C6.5/rGel fusions in xenograft models.
`We evaluated the ability of various C6.5/rGel fusion constructs to
`inhibit the growth of established SKOV3 tumor xenografts in nude
`mice after systemic administration. Tumors were induced in nude
`mice by s.c. injection of SKOV3 cells on day 0, and treatment was
`initiated on day 9 post-injection when the tumors were well estab-
`
`lished. Treatment consisted of five i.v. injections every other day.
`Groups of mice were treated at doses of 40 and 20 mg/kg for each
`fusion construct. Control mice were treated with PBS or 20 mg/kg
`rGel only. As shown in Fig. 5A and B, treatment with C6.5-L-rGel
`exhibited a significant antitumor effect. Mice treated with the 40
`mg/kg dose of C6.5-L-rGel showed a long-lasting antitumor effect
`that lasted more than 1 month until the animals were sacrificed. In
`mice treated with the 20 mg/kg dose, tumor growth was, in most
`cases, arrested for the duration of the treatment and resumed a
`couple of weeks after its completion. Otherwise, treatment of mice
`with 40 mg/kg C6.5-Fpe-rGel resulted in a significant delay in tu-
`mor growth. This was similar to the effect observed with the same
`dose of C6.5-L-rGel, but no significant effect could be observed at
`the lower (20 mg/kg) dose level. In contrast, mice treated with
`C6.5-Fdt-rGel at either dose (40 or 20 mg/kg) showed no specific
`antitumor effect above that observed with rGel alone.
`
`Figure 4. Western blot analysis of the
`cell killing mechanism of C6.5/rGel
`immunotoxins on SKOV3 cells. A, analysis
`of PARP cleavage after 24 and 48 h of
`C6.5/rGel fusion treatment. B, analysis of
`LC3 after treatment with C6.5/rGel fusions.
`The histogram shows quantitation of the
`ratio of LC3-II to β-actin. C, analysis of cell
`extract and medium for HMGB1 protein
`after C6.5/rGel treatment for 48 h.
`
`Cancer Res 2009; 69: (23). December 1, 2009
`
`8992
`
`www.aacrjournals.org
`
`Downloaded from
`
`on October 7, 2014. © 2009 American Association for Cancercancerres.aacrjournals.org
`
`
`Research.
`
`IMMUNOGEN2111, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Published OnlineFirst November 24, 2009; DOI: 10.1158/0008-5472.CAN-09-2693
`
`Novel Immunotoxins Targeting Her2/neu
`
`with reduced immunogenicity without affecting efficacy, toxicity,
`or specificity have not generally focused on anti-Her2/neu agents
`(26, 38, 39). To the best of our knowledge

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