throbber
C H A P T E R E I G H T
`
`Cell-Targeting Fusion Constructs
`Containing Recombinant Gelonin
`
`Mi-Ae Lyu, Yu (Joshua) Cao, Khalid A. Mohamedali, and
`Michael G. Rosenblum
`
`Contents
`1. Introduction
`2. Engineered Proteins Targeting Her2/neu
`2.1. Background
`2.2. Results
`2.3. Translational relevance
`3. rGel/BLyS Targeting Malignant B-Cells
`3.1. Background
`3.2. Results
`3.3. Translational relevance
`4. VEGF121/rGel Targeting Tumor Vasculature and Skeletal Metastases
`4.1. Receptor specificity of VEGF121/rGel by ELISA
`4.2. Cytotoxicity of VEGF121/rGel fusion toxin
`4.3. Internalization of VEGF121/rGel
`4.4. Angiogenesis assessment in chicken chorioallantoic
`membranes
`4.5. Targeting of VEGF121/rGel to blood vessels of orthotopic
`tumors in mice
`4.6. In vivo efficacy of VEGF121/rGel against solid tumors in nude
`mice
`4.7. In vivo efficacy of VEGF121/rGel against pulmonary
`metastases of MDA-MB-231 breast tumors
`4.8. In vivo efficacy of VEGF121/rGel against PC-3 prostate tumor
`growth in bone
`4.9. VEGF121/rGel inhibits the osteoblastic growth of MDA PCa
`118b cells in bone and normalizes the bone volume of 118b-
`tumor-containing bone
`4.10. Translational relevance
`References
`
`168
`169
`169
`171
`181
`181
`181
`183
`194
`195
`196
`197
`198
`
`199
`
`200
`
`202
`
`204
`
`206
`
`207
`208
`210
`
`Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental Therapeutics,
`M.D. Anderson Cancer Center, Houston, Texas, USA
`
`Methods in Enzymology, Volume 502
`ISSN 0076-6879, DOI: 10.1016/B978-0-12-416039-2.00008-2
`
`# 2012 Elsevier Inc.
`All rights reserved.
`
`167
`
`IMMUNOGEN 2113, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`168
`
`Mi-Ae Lyu et al.
`
`Abstract
`Therapeutic agents capable of targeting tumor cells present as established
`tumors and micrometastases have already demonstrated their potential in
`clinical trials. Immunotoxins targeting hematological malignancies and solid
`tumors have additionally demonstrated excellent clinical activity. This review
`focuses on our design and characterization studies of constructs composed of
`recombinant gelonin toxin fused to either growth factors or single-chain anti-
`bodies targeting solid tumor cells, tumor vasculature or hematological malig-
`nancies. These agents demonstrate cytotoxicity at nanomolar or sub-nanomolar
`levels. All of these constructs display impressive selectivity and specificity for
`antigen-bearing target cells in vitro and in vivo and are excellent clinical trial
`candidates.
`
`1. Introduction
`
`Although they are capable of selectively binding to cancer cells, most
`antibodies and other cell-targeting therapeutic agents alone have only
`modest antitumor properties primarily through ADCC activation, direct
`tumor cell signaling generating apoptosis or interference with essential
`growth factor homeostatic mechanisms. The majority of these agents have
`defined clinical utility primarily when used in combination with chemo-
`therapy or radiation therapy or as a part of a regimen using all three
`modalities. In order to improve the effectiveness of targeted therapeutic
`molecules, numerous groups have developed highly cytotoxic payloads
`conjugated or fused to these cell-targeting molecules. The concept is that
`the cell-targeting component serves to direct the agent to tumor cells in the
`body and to internalize into the cells themselves once they arrive. This
`specialized entry route also internalizes the attached cytotoxic payload
`directly into the tumor cells sparing normal tissues.
`There is now significant scientific and commercial interest in the devel-
`opment and application of antibody-directed cytotoxic molecules such as
`antibody-drug conjugates (ADCs) (Alley et al., 2010; Hughes, 2010; Mayes
`et al., 2011; Teicher, 2009; Webb, 2011).These constructs have gained
`considerable attention recently based on several promising clinical trial
`results demonstrating efficacy against a number of tumor targets (Kovtun
`and Goldmacher, 2007; Morrow et al., 2009; Murphy and Modi, 2009;
`Polson and Sliwkowski, 2009; Polson et al., 2009; Senter, 2009). It is
`important to note that the recent positive results with ADCs come as a
`second “Golden Age” well after notable disappointing clinical results of
`conjugates produced in the 1980s.
`The use of protein toxins as payloads linked to cell-targeting molecules is
`an interesting analogous approach to ADC technology. There have been a
`
`IMMUNOGEN 2113, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Cell-Targeting Fusion Constructs Containing Recombinant Gelonin
`
`169
`
`number of highly cytotoxic protein payloads developed including ricin
`toxin A-chain (RTA) (Kreitman, 2001; Lord et al., 2003; Wawrzynczak
`and Derbyshire, 1992), saporin (Flavell, 1998), pseudomonas exotoxin A
`(PE) (Kreitman, 2009; Pastan, 2003; Wolf and Elsasser-Beile, 2009), diph-
`theria toxin (DT) (Coll-Fresno et al., 1997; Hertler and Frankel, 1989; Negro
`and Skaper, 1997), and gelonin (Hertler and Frankel, 1989; Huang et al.,
`2010; Kwon et al., 2008; Sandvig and van Deurs, 2005). More recently,
`several groups (Hursey et al., 2002; Newton et al., 2001) including ours
`(Kurschus and Jenne, 2010; Kurschus et al., 2004; Rosenblum and Barth,
`2009; Stahnke et al., 2008) have developed completely human cytotoxic
`proteins for use as payloads in an attempt to circumvent concerns regarding
`the development of immunogenicity during long-term use with these agents.
`The highly cytotoxic plant toxin gelonin was first identified by Stirpe
`et al. (1980) as an excellent payload for the development of immunotoxins.
`This protein was identified as a new member of a class of ribosome-
`inactivating proteins (RIPs) with n-glycosidase activity similar to that of
`RTA. The crystal structure of gelonin has been reported (Hosur et al., 1995;
`Satyamurthy et al., 1994). A number of groups have utilized this molecule
`for generation of chemical conjugates (Chu et al., 2006; Fishwild et al.,
`1994; Harris et al., 1991; McGraw et al., 1994; McIntyre et al., 1994; Mehta
`et al., 2004; Mujoo et al., 1991; Pagliaro et al., 1998; Rosenblum et al., 1991,
`1992, 1999; Schwartz et al., 1987). In 1995, two separate groups reported
`the cloning and sequencing of a gene encoding the gelonin protein (Nolan
`et al., 1993; Rosenblum et al., 1995) and described a recombinant version
`(rGel) with enzymatic activity virtually identical to the native material. The
`recombinant version of the toxin has been employed by a number of groups
`as an excellent, flexible fusion partner (Cao et al., 2009; Lyu et al., 2007;
`Nimmanapalli et al., 2007; Veenendaal et al., 2002) allowing the construc-
`tion of fusion proteins at either the N-terminus or C-terminus of the rGel
`molecule.
`In this chapter, we will review some of the unique features—including
`the molecular events, unique signaling events, specific cytotoxicity, and
`in vivo studies of three fusion constructs we have generated which contain
`the toxin rGel.
`
`2. Engineered Proteins Targeting Her2/neu
`
`2.1. Background
`
`The Her2/neu proto-oncogene encodes a 185kDa transmembrane glyco-
`protein kinase with extensive homology to the epidermal growth factor
`receptor (EGFR, HER1). Amplification of the gene and overexpression of
`the Her2/neu protein product on tumor cells have been well described in
`
`IMMUNOGEN 2113, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`170
`
`Mi-Ae Lyu et al.
`
`numerous human cancers, including mammary and ovarian carcinomas, and
`gastric and lung tumors. Because Her2/neu plays a central role in malignant
`transformation and growth, it provides an attractive target for focused
`therapeutic approaches (Abramson and Arteaga, 2011; Cai et al., 2010;
`Mannocci et al., 2010).
`A number of approved immunotherapeutic agents directed at tumors
`which express high levels of Her2/neu, such as the monoclonal antibody
`trastuzumab (Herceptin) and small molecule inhibitors such as gefitinib
`(Iressa), have shown promising results, but the development of resistance
`to treatment remains a well-known problem (Garrett and Arteaga, 2011).
`To enhance its clinical potential, cell-surface Her2/neu has been targeted
`using ADCs or immunotoxins, composed of plant or bacterial toxins linked
`with a targeting molecule composed of monoclonal antibodies or antibody
`fragments (Govindan and Goldenberg, 2010; Isakoff and Baselga, 2011;
`Zielinski et al., 2009). Previously, a recombinant, murine anti-Her2/neu
`single-chain antibody (scFv) designated e23 has been fused to catalytic
`toxins such as Pseudomonas exotoxin A (PE) (Liu et al., 2009; Shinohara
`et al., 2002), to specifically target Her2/neu expressing cells. A major
`drawback of such proteins is their potential for immune response after
`repeated administration. Further complications could result from nonspe-
`cific binding of foreign proteins to vascular endothelial cells leading to
`vascular leak syndrome and ultimately interstitial edema and organ failure.
`The development of immunotoxins containing human or humanized
`components may circumvent these problems. Such immunotoxins may
`display reduced immunogenicity although antibodies to the toxin compo-
`nents may still
`limit prolonged therapy. We previously reported
`(Rosenblum et al., 1999) in vitro characterization and in vivo antitumor
`efficacy studies of an immunotoxin composed of the human chimeric
`anti-Her2/neu antibody (BACH-250) chemically conjugated to recombi-
`nant gelonin (rGel). rGel is a 29kDa ribosome-inactivating plant toxin with
`a potency and mechanism of action similar to RTA but with improved
`stability and reduced toxicity. The BACH-250/rGel conjugate demon-
`strated potent and specific cytotoxicity against Her2/neu overexpressing
`human tumor cells in culture and against SKOV3 tumor xenografts. How-
`ever, the treatment of solid tumors presents a potential problem since full-
`length antibodies must diffuse into the tumor against a hydrostatic pressure
`gradient and into disordered vasculature.
`Schier et al. (1995) and Adams et al. (1998) 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. Utilizing scFv C6.5,
`McCall et al. (1999) constructed and characterized a bispecific scFv com-
`posed of C6.5 and anti-CD16 scFv, displaying a high level of in vitro tumor
`cell cytotoxicity and in vivo tumor targeting. Studies by Park et al. (2001,
`2002) generated immunoliposomes containing doxorubicin which were
`
`IMMUNOGEN 2113, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Cell-Targeting Fusion Constructs Containing Recombinant Gelonin
`
`171
`
`targeted to tumor cells using antibody C6.5. These constructs demonstrated
`selective enhancement of the therapeutic index of doxorubicin chemother-
`apy. Most recently, Robinson et al. (Abramson and Arteaga, 2011; Adams
`et al., 2000; Cai et al., 2010; Mannocci et al., 2010; Robinson et al., 2008)
`successfully utilized a C6.5 diabody construct as a radioimmunotherapeutic
`agent containing (211At) for the treatment of Her2/neu positive solid
`tumors in xenograft models, demonstrating that scFv C6.5 could be utilized
`effectively in vehicles for targeted radioimmunotherapy by using powerful,
`short-lived a-emitting radioisotopes.
`In the present study, we describe the construction and characterization of
`several rGel-based chimaeric toxins composed of the scFv e23 or C6.5 and
`employing various linker configurations to examine how different antibodies
`and linker choices impact the in vitro and in vivo efficacy of fusion constructs.
`
`2.2. Results
`2.2.1. Construction, expression, and purification of rGel-based
`fusions
`The initial
`rGel-based immunotoxins consisted of a flexible linker
`(GGGGS, “L”) tethering the C-terminus of the e23 or C6.5 to the native
`rGel N-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 incorporating two different
`enzymatically sensitive furin cleavage linkers between the scFv and rGel
`toxin components. The two furin sensitive sequences designated
`“Fpe” (TRHRQPRGWEQL) and “Fdt” (AGNRVRRSVG), respectively
`(Fig. 8.1A). Several biochemical studies have demonstrated that the serine
`protease furin efficiently cleaves proteins containing these recognition
`sequences.
`Following purification, all the rGel-based immunotoxins migrated on
`SDS-PAGE at the expected molecular weight of 55kDa (Fig. 8.1B). How-
`ever, 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 occurring precisely at the predicted furin
`cleavage site producing the 27–28kDa fragments of
`scFv and rGel.
`Further analysis indicated the yields for each protein (per liter of bacterial
`culture) were 1.55mg for e23-L-rGel, 1.05mg for C6.5-L-rGel, 1.08mg for
`C6.5-Fpe-rGel, and 0.70mg for C6.5-Fdt-rGel.
`
`2.2.2. Characterization of e23-L-rGel and C6.5-L-rGel immunotoxins
`2.2.2.1. Binding activity To ensure that immunotoxins retained antigen
`binding ability, the fusion proteins were compared in an ELISA-based
`binding assay (Fig. 8.2A) using Her2/neu positive SKOV3 and Her2/neu
`negative MCF7 cells. The equilibrium dissociation constant Kd was further
`
`IMMUNOGEN 2113, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`172
`
`Mi-Ae Lyu et al.
`
`rGel
`
`rGel
`
`rGel
`
`rGel
`
`L
`
`L F
`
`pe
`
`Fdt
`
`VL–VH
`
`VH–VL
`
`VH–VL
`
`VH–VL
`
`M arker 1
`
`M arker 2
`
`C 6.5-F dt-r G el
`C 6.5-F pe-r G el
`C 6.5-L-r G el
`e23-L-r G el
`
`A
`e23-L-rGel
`
`C6.5-L-rGel
`
`C6.5-Fpe-rGel
`
`C6.5-Fdt-rGel
`
`B
`
`kDa
`
`150
`100
`75
`
`50
`
`35
`
`25
`
`Figure 8.1 Preparation of e23/rGel and C6.5/rGel series immunotoxins. (A) Sche-
`matic 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.
`
`¼8.5
`calculated (Graphpad Prism, V4.03). The affinity of e23-L-rGel (Kd
`¼12.6 nM).
`nM) for SKOV3 cells was similar to that for C6.5-L-rGel (Kd
`The Kd values were consistent with those previously measured in an in vitro
`live cell assay using scFv itself. In addition, both immunotoxins demon-
`strated significant specificity based on the background of binding to MCF7
`cells. ELISA assay suggested that the human scFv C6.5 displayed similar
`binding specificity compared to the murine e23.
`
`2.2.2.2. 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 incorporation by
`isolated
`rabbit
`reticulocytes.
`Inhibition
`curves
`for
`the
`fusion
`constructs e23-L-rGel, C6.5-L-rGel, and native rGel were compared
`(Fig. 8.2B), and IC50 values for the three molecules were found to be
`
`IMMUNOGEN 2113, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Cell-Targeting Fusion Constructs Containing Recombinant Gelonin
`
`173
`
`SKOV3
`
`MCF7
`
`e23-L-rGel
`C6.5-L-rGel
`rGelonin
`
`0
`
`10
`20
`30
`40
`Concentration (nM)
`
`50
`
`60
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Absobance at 405 nm
`
`e23-L-rGel
`C6.5-L-rGel
`rGelonin
`
`0
`
`30
`20
`40
`10
`Concentration (nM)
`
`50
`
`60
`
`A
`
`0.8
`
`0.6
`
`0.4
`
`0.2
`
`0.0
`
`Absobance at 405 nm
`
`e23-L-rGel
`
`C6.5-L-rGel
`
`rGelonin
`
`C
`
`SKOV3
`
`MCF7
`
`e23-L-rGel, IC50= 15.4 pM
`C6.5-L-rGel, IC50= 15.5 pM
`rGelonin, IC50= 10.6 pM
`
`10-13
`
`10-12
`10-11
`10-10
`Concentration (M)
`
`10-9
`
`10-8
`
`B
`
`120
`
`100
`
`80
`
`60
`
`40
`
`20
`
`Percent control
`
`0
`10-14
`
`Figure 8.2 Characterization and comparison of e23-L-rGel and C6.5-L-rGel immu-
`notoxins. (A) Evaluation 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 (RRLA).
`(C) Internalization of e23-L-rGel and C6.5-L-rGel on SKOV3 and MCF7 cells.
`Cells were subjected to immunofluorescent staining with anti-rGel antibody (FITC-
`conjugated secondary), with propidium iodine nuclear counterstaining.
`
`virtually identical (15.41, 15.52 vs. 10.6 pM, respectively), suggesting that
`no loss of toxin activity occurred in the fusion molecules.
`
`2.2.2.3. Cellular uptake of immunotoxins We next examined whether
`the e23-L-rGel and C6.5-L-rGel fusions could specifically internalize into
`target cells. Immunofluorescence staining was performed on SKOV3
`and MCF7 cells after exposure to the constructs. As shown in Fig. 8.2C,
`the rGel moiety of both fusions was observed primarily in the cytosol
`after treatment of SKOV3, but not in MCF7 cells, demonstrating that
`both constructs were comparable in efficient cell binding and rapid
`internalization after exposure to Her2/neu positive cells.
`
`2.2.2.4. In vitro cytotoxicity The e23-L-rGel, C6.5-L-rGel constructs
`and rGel were tested against a number of different tumor cell
`lines
`(Table 8.1). The SKBR3 cells with the highest level of Her2/neu expres-
`sion were killed most efficiently by both antibody-fusion constructs, with
`
`IMMUNOGEN 2113, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Table 8.1 Comparative IC50 values of e23-L-rGel and C6.5-L-rGel fusion constructs against various types of tumor cell lines
`
`Cell line
`
`Origin
`
`Her2/neu level
`
`IC50 (nM)
`
`Targeting index
`
`a
`
`SKBR3
`NCI-N87
`Calu3
`SKOV3
`BT474
`MDA MB435S
`MCF7
`4T1
`
`High
`Human, breast
`High
`Human, gastric
`High
`Human, lung
`Human, ovarian High
`Human, breast
`High
`Human, breast
`Medium
`Human, breast
`Low
`Mouse, breast
`No
`
`a Targeting index represents IC50 of rGel/IC50 of immunotoxin.
`
`e23-L-rGel
`
`C6.5-L-rGel
`
`rGel
`
`e23-L-rGel
`
`C6.5-L-rGel
`
`6.0
`59.2
`41.1
`16.3
`27.1
`24.6
`266.3
`>1000
`
`9.1
`45.0
`31.3
`18.0
`25.2
`28.8
`200.9
`>1000
`
`1671.0
`1334.0
`879.7
`378.9
`325.2
`359.0
`260.4
`>1000
`
`279
`23
`21
`23
`12
`15
`1
`1
`
`184
`30
`28
`21
`13
`12
`1
`1
`
`IMMUNOGEN 2113, pg. 8
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Cell-Targeting Fusion Constructs Containing Recombinant Gelonin
`
`175
`
`IC50 values of 6.0 and 9.1 nM for e23-L-rGel and C6.5-L-rGel, respec-
`
`tively. IC50 values for rGel toxin were 200-fold higher (1671nM). For the
`
`other Her2/neu positive cells, both immunotoxins also showed similar IC50
`values demonstrating that the two fusion proteins possess very similar cell-
`killing activity and specificity. Further, the MCF7 and 4T1 cells which
`express relatively low levels of Her2/neu demonstrated little to no specific
`cytotoxicity of the fusion constructs compared to rGel itself, clearly demon-
`strating that the presence of higher levels of cell-surface Her2/neu is
`required for specific cytotoxicity of the constructs.
`
`2.2.2.5. 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 constructs. 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 chi-
`maeric toxins to proteolytic cleavage, purified fusions were subjected to
`proteolysis with recombinant furin (Fig. 8.3A). 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 efficiency
`(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 constructs. 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.
`
`2.2.2.6. 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 (Table 8.2). Interestingly, the cell lines
`showed no differences in overall sensitivity to the fusion constructs with the
`incorporation of furin cleavage linkers compared with flexible L linker. All
`the fusions showed potent cytotoxicity after 48h and exerted highly potent
`cell-killing at 72h. This suggests that the cleavage efficiency of different
`linkers for these chimaeric toxins was not a major determinant to the overall
`cytotoxic effects significant observed on different linkers. Surprisingly, the
`cytotoxic kinetics of the constructs therefore appeared to be independent of
`the sensitivity of the constructs to proteolytic cleavage.
`
`2.2.2.7. Intracellular
`from various constructs The
`rGel
`release of
`intracellular release of rGel after endocytosis of various C6.5/rGel fusion
`constructs was assessed by Western blot with an anti-rGel antibody
`(Fig. 8.3B). During the treatment of SKOV3 cells, rGel release was found
`
`IMMUNOGEN 2113, pg. 9
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Mi-Ae Lyu et al.
`
`B
`
`C6.5-L-rGel
`
`C6.5-Fpe-rGel
`
`C6.5-Fdt-rGel
`
`kDa
`
`1 2 4 8 12 24 h
`
`1 2 4 8 12 24 h 1 2 4 8 12 24 h
`
`C6.5-L-rGel
`
`C6.5-Fpe-rGel C6.5-Fdt-rGel
`
`pH 5.4
`pH 7.2
`uncut
`
`pH 5.4
`pH 7.2
`uncut
`
`pH 5.4
`pH 7.2
`uncut
`
`0 h
`24 h
`48 h
`72 h
`
`C6.5-L-rGel
`C6.5-Fpe-rGel C6.5-Fdt-rGel
`rGel-based fusions
`
`58
`46
`
`30
`25
`
`b-actin
`
`60
`50
`40
`30
`20
`10
`0
`
`IC50 (nM)
`
`18.5 4.5 5.8 100 100
`
`C6.5-L-rGel
`C6.5-Fpe-rGel
`C6.5-Fdt-rGel
`
`0
`
`10
`
`20
`30
`40
`50
`60
`Incubation time (h)
`
`70
`
`80
`
`176
`
`A
`
`kDa
`
`58
`46
`
`30
`25
`
`Cleavage
`rate (%)
`
`100
`
`80
`
`60
`
`40
`
`20
`
`0
`
`C
`
`% Maximal binding
`
`Figure 8.3 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) Func-
`tional stability analysis of the fusions by whole-cell ELISA and cytotoxicity on SKOV3
`
`C for up to 72h before test.
`cells. The proteins were incubated in human plasma at 37
`
`Table 8.2 Kinetics of cytotoxicity of rGel-based immunotoxins
`
`IC50 (nM)
`
`SKBR3
`
`SKOV3
`
`Calu3
`
`MDA MB435S
`
`24
`─a
`─
`─
`
`48
`
`30.1
`36.4
`44.3
`
`72
`24
`48
`9.4 ─ 102.3
`9.3 ─ 175.1
`10.4 ─ 203.2
`
`72
`24
`48
`26.6 ─ 250.5
`34.2 ─ 185.8
`30.9 ─ 468.8
`
`72
`24
`48
`31.9 ─ 251.7
`27.5 ─ 260.1
`88.1 ─ 277.4
`
`72
`
`25.3
`25.9
`30.5
`
`Toxins duration (h)
`
`C6.5-L-rGel
`C6.5-Fpe-rGel
`C6.5-Fdt-rGel
`
`a –, not detected.
`
`to be maximal at 2h after treatment with C6.5-L-rGel and 4h after exposure
`to C6.5-Fpe-rGel. For C6.5-Fdt-rGel, the rGel component was released
`within 1–2h 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 inter-
`nalization. Although the maximal rGel release of different fusions was
`achieved at different time points, the absolute amounts of delivered rGel
`
`IMMUNOGEN 2113, pg. 10
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Cell-Targeting Fusion Constructs Containing Recombinant Gelonin
`
`177
`
`were virtually identical. Therefore, this data confirms the observation that
`introduction of an unstable furin cleavage linker does not improve the
`intracellular rGel release of the constructs.
`
`2.2.2.8. Functional stability analysis of C6.5/rGel fusions The linkers
`between C6.5 and rGel demonstrated a differential sensitivity to protease
`action which may result in different clearance and metabolic kinetics in vivo
`(Clemente and de la Torre, 2007; Zhang et al., 2008). 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. 8.3C). Our results showed
`that in the presence of human plasma, the C6.5-Fdt-rGel construct dis-
`played a reduction in binding activity within 6h of incubation and a 20%
`loss of binding activity after 72h incubation. In contrast, C6.5-L-rGel
`and C6.5-Fpe-rGel fusion constructs demonstrated only a 9% and 12%
`reduction, respectively, after 72h incubation.
`In addition, the immunotoxins were evaluated for cytotoxic activity fol-
`lowing incubation in human plasma for 0, 24, 48, and 72h (Fig. 8.3C). For the
`C6.5-Fdt-rGel construct, the cell-killing activity was reduced over twofold
`after 48h as indicated by increasing IC50 values of 20 nM versus 48 nM.
`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 48h, and displayed a little
`influence on IC50 after 72h incubation in plasma (16 nM vs. 22 nM and 17 nM
`vs. 25 nM for each construct). This functional stability analysis indicated that
`compared with L and Fpe linker, the Fdt linker was much more unstable in
`human plasma and this may reduce the in vivo potency of potential therapeutic
`applications using constructs containing this linker design.
`
`2.2.2.9. Mechanistic studies 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. 8.4A,
`C6.5/rGel fusions did not demonstrate activation of caspase-dependent
`apoptosis in SKOV3 cells and showed no cleavage of caspase substrate
`PARP. The TUNEL results (Fig. 8.4B) confirmed that the cytotoxic effects
`of the rGel-based fusions were not mediated by apoptosis and DNA
`fragmentation.
`To assess whether necrotic cell death was induced, we examined LDH
`release which is a marker of abrupt membrane lysis. In this case, treatment of
`SKOV3 cells with Triton X-100 serves as a positive control causing LDH
`release (Fig. 8.5). In contrast, treatment with the fusion constructs failed to
`demonstrate LDH release indicating that the observed cytotoxicity did not
`appear to be the result of necrosis.
`
`IMMUNOGEN 2113, pg. 11
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`178
`
`A
`
`24 h
`
`PARP
`
`b-actin
`
`B
`
`C 6.5-F pe-rG el
`C 6.5-F dt-rG el
`C 6.5-L-rG el
`
`-
`
`rG el
`
`48 h
`
`PARP
`
`b-actin
`
`Mi-Ae Lyu et al.
`
`C 6.5-F pe-rG el
`C 6.5-F dt-rG el
`C 6.5-L-rG el
`
`-
`
`rG el
`
`No treatment
`
`DNase I
`
`rGel
`
`C6.5-L-rGel
`
`C6.5-Fpe-rGel
`
`C6.5-Fdt-rGel
`
`Figure 8.4 Apoptosis analysis of C6.5/rGel immunotoxins on SKOV3 cells. (A) Anal-
`ysis of PARP cleavage after 24 and 48h of C6.5/rGel fusions treatment. (B) Assessment
`of apoptosis in SKOV3 cells treated with rGel-based immunotoxins by TUNEL assay.
`SKOV3 cells were treated with 25 nM of C6.5/rGel fusions or rGel for 72h, then
`stained for DNA fragmentation. As shown, neither the C6.5/rGel fusions nor the rGel
`itself affected DNA fragmentation.
`
`48 h
`
`100
`
`75
`
`50
`
`25
`
`LDH release (%)
`
`0
`
`Triton X-100
`
`-
`
`rGel
`
`C6.5-Fdt-rGel
`C6.5-Fpe-rGel
`C6.5-L-rGel
`
`24 h
`
`100
`
`75
`
`50
`
`25
`
`0
`
`LDH release (%)
`
`Triton X-100
`
`-
`
`rGel
`
`C6.5-L-rGel
`C6.5-Fdt-rGel
`C6.5-Fpe-rGel
`
`Figure 8.5 Evaluation of necrosis by LDH release in the SKOV3 cells after treatment
`with C6.5/rGel fusions or Triton X-100; no LDH is released in the fusion-treated cells
`(meanS.D. from three replicates).
`
`IMMUNOGEN 2113, pg. 12
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Cell-Targeting Fusion Constructs Containing Recombinant Gelonin
`
`179
`
`C 6.5-L-r G el
`
`C 6.5-F pe-r G el
`
`C 6.5-F dt-r G el
`
`r G el
`
`-
`
`r G el
`
`C 6.5-F dt-r G el
`C 6.5-F pe-r G el
`C 6.5-L -r G el
`
`48 h
`
`LC3-I
`LC3-II
`
`b-actin
`
`2.0
`
`1.5
`
`1.0
`
`0.5
`
`0.0
`
`LC3-II/b-actin ratio
`
`C 6.5-L-r G el
`
`C 6.5-F pe-r G el
`
`C 6.5-F dt-r G el
`
`r G el
`
`-
`
`r G el
`
`C 6.5-F dt-r G el
`C 6.5-F pe-r G el
`C 6.5-L -r G el
`
`Cells
`
`C 6.5-L-r G el
`
`C 6.5-F pe-r G el
`
`C 6.5-F dt-r G el
`
`r G el
`
`Medium
`
`C 6.5-L-r G el
`
`C 6.5-F pe-r G el
`
`C 6.5-F dt-r G el
`
`r G el
`
`A
`
`24 h
`
`LC3-I
`LC3-II
`
`b-actin
`
`2.0
`
`1.5
`
`1.0
`
`0.5
`
`0.0
`
`LC3-II/b-actin ratio
`
`B
`
`HMGB1
`b-actin
`
`Figure 8.6 Western blot analysis of cell-killing mechanism of C6.5/rGel immunotox-
`ins on SKOV3 cells. (A) Analysis of PARP cleavage after 24 and 48h of C6.5/rGel
`fusions treatment. (B) Analysis of LC3 after treated with C6.5/rGel fusions. The
`the ratio of LC3-II compared with b-actin.
`histogram shows quantitation of
`(C) Analysis of cell extract and medium for HMGB1 protein after C6.5/rGel treatment
`for 48h.
`
`the immunotoxins activate autophagic signaling
`We next asked if
`in SKOV3 cells. MAP LC3-I, known to be usually present in the cytosol, is
`palmitoylated during autophagy to form membrane-bound LC3-II and
`is associated with autophagosomes. As shown in Fig 8.6A, the ratio of
`LC3-II formation to the b-actin control was increased after treatment with
`the fusion constructs demonstrating 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. 8.6B). Tumor cells that are dying with autophagy selectively
`release the nuclear HMGB1 protein, without displaying characteristics of
`necrosis. These data indicated that the observed cytotoxic effects of C6.5/rGel
`fusions in SKOV3 cells appeared to be mediated not through an apoptotic
`or necrotic mechanisms but by the efficient induction of autophagic cell death.
`
`C6.5/rGel
`2.2.2.10. Antitumor
`xenograft
`in
`fusions
`of
`activity
`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.
`
`IMMUNOGEN 2113, pg. 13
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Mi-Ae Lyu et al.
`
`PBS
`rGelonin (20 mg/kg)
`
`C6.5-L-rGel (20 mg/kg)
`C6.5-Fpe-rGel (20 mg/kg)
`
`C6.5-Fdt-rGel (20 mg/kg)
`
`1500
`
`1250
`
`1000
`
`750
`
`500
`
`250
`
`B
`
`Tumor volume (mm3)
`
`PBS
`
`rGelonin (20 mg/kg)
`
`C6.5-L-rGel (40 mg/kg)
`
`C6.5-Fpe-rGel (40 mg/kg)
`
`C6.5-Fdt-rGel (40 mg/kg)
`
`180
`
`1500
`
`1250
`
`1000
`
`750
`
`500
`
`250
`
`A
`
`Tumor volume (mm3)
`
`0
`
`0
`
`10
`
`20
`
`30
`
`40
`50
`Time (days)
`
`0
`
`0
`
`10
`
`20
`
`30
`
`40
`50
`Time (days)
`
`FITC/rGel
`
`PI/nucleus
`
`C
`
`C6.5-L-rGel
`
`rGelonin
`
`Figure 8.7 The antitumor activity of C6.5/rGel immunotoxins against SKOV3 tumor
`xenografts in nude mice. (A, B) Treatments of SKOV3 flank tumors with C6.5/rGel
`fusions at the dose of 40 and 20mg/kg. Antitumor effects of i.v. injections of PBS, rGel,
`C6.5-L-rGel, C6.5-Fpe-rGel, or C6.5-Fdt-rGel on SKOV3 tumors. Mean tumor vol-
`
`ume was calculated by WLH as measured by digital calipers. (C) Immunofluores-
`
`cence staining of tumor samples after i.v. injection of C6.5-L-rGel and rGel. 24h after
`injection, animal was sacrificed and frozen tumor sections were prepared and detected
`by anti-rGel antibody. Propidium iodide was used for DNA staining.
`
`injection of SKOV3 cells on day 0, and treatment was initiated on day 9
`postinjection when the tumors were well-established. Treatment consisted
`of five i.v. injections every other day. Groups of mice were treated at doses
`of 40 and 20mg/kg for each fusion construct. Control mice were treated
`with PBS or 20mg/kg rGel only. As shown in Fig. 8.7A and B, treatment
`with C6.5-L-rGel exhibited a significant antitumor effect. Mice treated at
`the 40mg/kg dose of C6.5-L-rGel demonstrated a long-lasting antitumor
`effect which lasted more than 1month until the animals were sacrificed.
`With mice treated at the 20mg/kg dose level, 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 40mg/kg of
`
`IMMUNOGEN 2113, pg. 14
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Cell-Targeting Fusion Constructs Containing Recombinant Gelonin
`
`181
`
`C6.5-Fpe-rGel, resulted in a significant delay in tumor 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 (20mg/kg) dose level. In
`contrast, mice treated with either dose (40 or 20mg/kg) of C6.5-Fdt-rGel
`showed no specific antitumor effect above that observed with rGel alone.
`We next examined the localization of C6.5-L-rGel and rGel after
`administration to mice bearing SKOV3 tumors. Immunofluorescence
`staining confirmed that the C6.5-L-rGel localized specifically in tumor
`tissue, but no staining was observed in tumors after administration of rGel
`itself (Fig. 8.7C). This suggests that the fusion construct C6.5-L-rGel can
`effectively target tumor cells overexpressing Her2/neu in vivo and can
`demonstrate significant tumor growth suppressive effects in the absence of
`observable toxicity.
`
`2.3. Translational relevance
`
`In conclusion, we have designed and developed several novel immunotox-
`ins containing the human scFv C6.5 and toxin rGel. These agents exhibit
`efficient cytotoxicity for Her2/neu overexpressing tumor cells, and the
`human antibody appears to be virtually identical as an effective carrier of
`rGel toxin compares to the murine e23. The introduction of a furin
`cleavable linker between C6.5 and rGel did not result in improved intrac

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