throbber
Chapter 18
` Design, Development, and Characterization
`of Recombinant Immunotoxins Targeting
`HER2/neu
`
` Yu Cao and Michael G. Rosenblum
`
` Background
`
` The human epidermal growth factor receptor 2 (HER2), also known as ErbB2,
`c-erbB2, or HER2/neu, was initially discovered in 1985 by two independent labora-
`tories [ 1, 2 ] . HER2/neu is a 185 kDa (1,255 aa) transmembrane receptor encom-
`passing an intracellular tyrosine kinase domain and an extracellular ligand binding
`component [ 3– 5 ] . Extensive clinical studies have shown that overexpression of
`HER2/neu is found in 20–40% of patients with breast, ovarian, endometrial, gastric,
`bladder, prostate, and lung cancers. Studies clearly demonstrate that HER2/neu over-
`expression correlates with the prevalence of metastatic spread of many tumors and
`is generally considered to be a poor prognostic indicator [ 6– 9 ] .
` Since HER2/neu overexpression by tumor cells is quite speci fi c, therapies directed
`against this receptor have rapidly gained recognition for their selectivity and ef fi cacy
`in the clinical setting. While targeting of HER2/neu with humanized antibodies such
`as trastuzumab (Herceptin; Genentech) has proven to be an effective approach for the
`treatment of HER2/neu-overexpressing breast cancers, there are a signi fi cant number
`of patients with HER2/neu-positive tumors who do not respond or who acquire resis-
`tance to this therapy [ 10– 13 ] . Therefore, there is a need for novel therapeutic
`approaches using HER2/neu not only as a target for interfering with the growth fac-
`tor signaling component but also for receptor-mediated delivery of cytotoxic agents.
` Immunotoxins are a novel approach for the development of highly speci fi c,
`targeted agents and which generally employ a powerful class of protein toxins
` [ 14, 15 ] . These include plant toxins such as ricin [ 16– 25 ] , saporin [ 26– 29 ] , and
`gelonin [ 30– 32 ] , which inactivate ribosomes, and single-chain bacterial toxins
`
` Y. Cao • M. G. Rosenblum (*)
` Immunopharmacology and Targeted Therapy Laboratory, Department of Experimental
`Therapeutics , MD Anderson Cancer Center , Houston , TX 77030 , USA
`e-mail: mrosenbl@mdanderson.org
`
`G.L. Phillips (ed.), Antibody-Drug Conjugates and Immunotoxins: From Pre-Clinical
`Development to Therapeutic Applications, Cancer Drug Discovery and Development,
`DOI 10.1007/978-1-4614-5456-4_18, © Springer Science+Business Media New York 2013
`
`319
`
`IMMUNOGEN 2016, pg. 1
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`320
`
`Y. Cao and M.G. Rosenblum
`
` Diphtheria toxin (DT) [ 33 ] and Pseudomonas exotoxin (PE) [ 34– 43 ] , which ADP
`ribosylate elongation factor 2 (EIF2). Anti-HER2/neu immunotoxins have been
`created initially by chemically conjugating an antibody to a whole protein toxin
`or, for more selective activity, using a protein toxin devoid of its natural binding
`domain [ 19, 23, 30, 44 ] . Technical advances in antibody engineering now enable
`us to produce various antibodies or antibody fragments in Escherichia coli , and as
`a result, HER2/neu-speci fi c antibodies and engineered fragments thereof have
`been developed to deliver various toxins to HER2/neu-positive tumor cells [ 45–
` 51 ] . Various anti-HER2/neu immunotoxins which have been developed or are cur-
`rently under evaluation are described in Table 18.1 .
`
` Antibody–Drug Conjugates: Promise and Problems
`
` Antibody-based therapeutics is of growing signi fi cance for cancer therapy. To date,
`two of the most promising strategies to enhance the antitumor activity of antibodies
`are antibody–drug conjugates (ADCs) and antibodies (or fragments) chemically
`conjugated or genetically fused to various toxins (immunotoxins).
` One successful application of the ADC approach is Trastuzumab–DM1. This is
`a covalent conjugation of trastuzumab with the maytansinoid DM1—a highly toxic
`derivative of the antimitotic drug maytansine. The therapeutic potential of
`Trastuzumab–DM1 has been extensively investigated in both in vitro and in vivo
`models of trastuzumab sensitive and insensitive breast cancers [ 52– 56 ] . It has dem-
`onstrated remarkable activity in phase I and II studies in which it was given to
`patients harboring trastuzumab-insensitive breast tumors [ 57, 58 ] . Furthermore,
`several other ADCs using anti-HER2/neu antibodies have been developed and have
`shown potent antitumor activity [ 59, 60 ] . Despite successful reports, it is important
`to note that this strategy has some limitations. The fi rst is the limited reproducibility
`of chemical conjugation due to the fact that there are numerous coupling sites on an
`antibody molecule. Secondly, chemically modi fi ed antibodies have demonstrated a
`greater tendency to aggregate, especially when multiple drug molecules are conju-
`gated to a single antibody. Furthermore, it is challenging to remove remaining
`unconjugated antibodies from the ADC mixture. Finally, the emergence of multi-
`drug resistance (MDR and MRP) mechanisms in tumors from heavily treated
`patients may engender cross-resistance to ADCs.
` With the development of recombinant DNA technology, anti-HER2/neu immu-
`notoxins composed of antibodies (or fragments) and protein toxins have become a
`promising alternative approach for HER2/neu-positive tumors. Compared to the
`ADC approach, one attractive advantage of immunotoxins is that the targeting
`antibody and antitumor toxin can be produced directly as a single molecule, thus
`avoiding laborious chemical conjugation steps. In addition, the linkage between
`the toxin and targeting antibody is identical and exactly de fi ned in a given prepara-
`tion of recombinant immunotoxin, thereby promoting homogeneity of the fi nal
`product. Compared with chemical conjugates, genetically engineered immunotoxins
`
`IMMUNOGEN 2016, pg. 2
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`18 Design, Development, and Characterization...
`
`321
`
`reast cancer [ 45 ]
`reast cancer [ 47 ]
`olon cancer [ 44 ]
`
` C
`
` B
`
` B
`
`olon cancer [ 33 ]
`pidermoid cancer [ 40, 193 ]
`
` E
`
` C
`
`ecombinant protein
`ecombinant protein
`hemical conjugation
`
` C
`
`ecombinant protein
`ndirect bridge-linking
`
` R
`
` I
`
`reast cancer [ 35, 39, 151 ]
`
` B
`
`iposome-mediated
`
` L
`
`chemical conjugation
`
`pidermoid cancer [ 37, 46 ]
`
` E
`
`ecombinant protein
`
` R
`
`pidermoid cancer [ 41, 51 ]
`
` E
`
`ecombinant protein
`
` R
`
`cancer [ 49 ] , breast cancer [ 48 ]
`
`astric cancer [ 38, 43 ] , epidermoid
`
` G
`
`ecombinant protein
`
`astric cancer [ 152 ] , ovarian cancer [ 192 ]
`
` G
`
`ecombinant gene
`
`delivery
`
`cancer [ 42, 50, 124 ]
`schwannoma cancer [ 191 ] , breast
`gastric cancer [ 84, 123, 188– 190 ] ,
`4, 184 ] , lung cancer [ 185– 187 ] ,
`
` 3
` [
`
`
`cancer [ 40, 183 ] , prostate cancer
`
`varian cancer [ 36, 122 ] , epidermoid
`ung cancer [ 25 ]
`
` L
`
` O
`
`ecombinant protein
`hemical conjugation
`
`cancer [ 16, 18, 19 ] , gastric cancer [ 22 ]
`reast cancer [ 17, 20, 21, 23, 24 ] , ovarian
`varian cancer [ 28 ]
`reast cancer [ 26, 29 ] , Melanoma [ 27 ]
`varian cancer [ 31 ] , breast cancer [ 32 ]
`varian cancer [ 30 ]
`umor type
`
` O
`
` O
`
` B
`
` O
`
` B
`
` T
`
`hemical conjugation
`ndirect bridge-linking
`hemical conjugation
`ecombinant protein
`hemical conjugation
`roduction
`
` P
`
` C
`
` R
`
` C
`
` I
`
` C
`
` C
`
` R
`
` R
`
` R
`
` R
`
` R
`
`uman anti-HER2 scFv
`uman anti-HER2 scFv
`urine anti-HER2 mAb
`and anti-EpCAM)
`urine-bispeci fi c scFv (anti-HER2
`umanized anti-HER2 mAb
`
` H
`
` M
`
` M
`
` H
`
`enterotoxin
`taphylococcal
`
` S
`
`iptheria toxin
`
` D
`
` H
`
`acillus Cyt2Aa1 toxin
`
` B
`
`umanized anti-HER2 Fab ¢
`
` H
`
`and anti-EGFR)
`urine-bispeci fi c scFv (anti-HER2
`(dsFv) 2
`urine bivalent anti-HER2 dsFv
`Fv fragments (dsFv)
`urine anti-HER2 disul fi de-stabilized
`
` M
`
` M
`
` M
`
`urine anti-HER2 scFv
`urine anti-HER2 Fab ¢
`
` M
`
` M
`
`seudomonas exotoxin
`
` P
`
`acteria
`
` B
`
` H
`
` M
`
` M
`
` M
`
`icin
`
` R
`
`aporin
`
` S
`
`lant
`oxin source
`able. 18.1 Immunotoxins developed for HER2/neu targeted therapy
`
`elonin
`oxin
`
` T
`
` G
`
` T
`
` T
`
` P
`
`urine anti-HER2 mAb
`urine anti-HER2 mAb
`urine anti-HER2 mAb
`uman anti-HER2 scFv
`umanized anti-HER2 mAb
`argeting device
`
` T
`
` H
`
`IMMUNOGEN 2016, pg. 3
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`322
`
`Y. Cao and M.G. Rosenblum
`
`can be easily designed to enhance antitumor ef fi cacy. Finally, data suggests that the
`emergence of MDR cellular protection mechanisms in heavily pretreated patients
`may not impact cytotoxic effects of immunotoxins.
` We have provided general principles for development of anti-HER2/neu immu-
`notoxins, and current strategies to employ these molecules for directed cancer ther-
`apy are discussed focusing mainly on design optimization to improve antitumor
`ef fi cacy and off-target toxicity.
`
` Anti-HER2/neu Immunotoxins
`
` Construction of Recombinant Immunotoxins
`
` HER2/neu-overexpressing cancers are a model of disease for the development of
` rationally designed targeted therapies. The scienti fi c advances in understanding the
`role of HER2/neu function, the structural aspects of HER2/neu function, and the sig-
`naling partners and circuitry underlying tumorigenic HER2/neu signaling have afforded
`unique opportunities for rational drug design to target these pathways. The develop-
`ment and application of various HER2/neu-targeted therapies has bene fi ted greatly by
`a more advanced understanding of HER2/neu function and biology [ 61, 62 ] .
` Overall, strategies to enhance anti-HER2/neu immunotoxin potency include
`improvements to the af fi nity and speci fi city of targeting moiety, identi fi cation and
`incorporation of new and better toxins, reengineering known toxins for reduced
`immunogenicity, and designing novel linkers between toxins and targeting moieties
`to optimize toxin translocation to the cytosol [ 63– 66 ] . Numerous excellent reviews
`have previously compared the advantages and disadvantages of a variety of cyto-
`toxic proteins including bacterial, plant, and mammalian toxins successfully
`employed for the construction of immunotoxins [ 67, 68 ] . This review will address
`how linker design and antibody af fi nity affect immunotoxins in tumor-speci fi c tar-
`geted therapies.
`
` Peptide Linker Designs
`
` The development of various linkers which bridge disparate molecules such as small
`drugs conjugated to tumor-targeting carriers has been the subject of numerous stud-
`ies for the past few years [ 69, 70 ] . Based on numerous prior studies, the incorpora-
`tion and design of linkers is critical to the success of ADCs. Conceptually, an ideal
`linker must be stable in systemic circulation, while being ef fi ciently cleaved to allow
`rapid release of an active form of the drug once the construct has been internalized
`into the tumor cell target. To this end, a variety of linkers have been designed with
`different chemical structures and stabilities [ 71, 72 ] . Selection of an appropriate
`linker depends on the type of cancer and the required cytotoxic agent. Furin is a
`
`IMMUNOGEN 2016, pg. 4
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`s==a-c:==o
`~;;;;;;;;;~~==·
`
`~:=.-..c:===·
`~=..-..:====·
`
`18 Design, Development, and Characterization...
`
`323
`
`
`
`a
`C6.5-L-rGel
`
`C6.5-Fpe-rGel
`
`C6.5-Fdt-rGel
`
`C6.5
`
`C6.5
`
`C6.5
`
`L
`
`Fpe
`
`Fdt
`
`rGel
`
`rGel
`
`rGel
`
`b
`C6.5
`
`ML3-9
`
`MH3-B1
`
`B1D3
`
`SSSN------YFQH
`
`SSSN------YFQH
`
`TDRT------YFQH
`
`VH
`
`VH
`
`VH
`
`VH
`
`A--DS
`
`S--YT
`
`S--YT
`
`VL
`
`VL
`
`VL
`
`VL
`
`Kd
`16nM
`
`1nM
`
`0.12nM
`
`0.013nM
`
`TDRT------WLDN
`
`S--YT
`
`c
`
`C6.5/ML3-9/MH3-B1/B1D3-rGel cDNA
`
`scFv
`
`L
`
`rGel
`
`BspHI
`
`XhoI
`
`NcoI
`
`pET 32a
`(Novagen)
`
`Bacterial Expression Vector
`
` Fig. 18.1 Construction and preparation of scFv/rGel immunotoxins. ( a ) Schematic diagram of
`immunotoxin constructs containing scFv C6.5, peptide linker (L, Fpe or Fdt), and rGel toxin. ( b )
`Amino acid mutations and af fi nity parameters of the C6.5 and its mutants, ML3-9, MH3-B1, and
`B1D3. The listed amino acids for each scFv indicate mutations to the sequence and the substituting
`amino acids. Dashes indicate no changes from the original sequence. ( c ) Diagram of immunotoxin
`constructions containing scFv (C6.5, ML3-9, MH3-B1, or B1D3) and rGel
`
`cellular endoprotease and has been implicated in proteolytically activating large
`numbers of secreted proteins such as prohormones, growth factors, receptors, and
`viral glycoproteins. These proteins are synthesized as inactive precursors and must
`be proteolytically cleaved to become functionally mature. In previous studies, the
`inclusion of furin-cleavable linkers into fusion constructs containing ribotoxins,
`caspase3, or granzyme B (GrB) has demonstrated a signi fi cant improvement in
`speci fi c toxicity compared to constructs containing stable linkers [ 73, 74 ] .
` The incorporation of cleavable linkers for immunotoxins is essential since, in
`general, the toxin components are enzymatically inactive in the construct until
`intracellular release from their cell-targeting carriers [ 75, 76 ] . For recombinant
`gelonin (rGel)-based constructs, the enzymatic ( N -glycosidase) activity of the toxin
`is preserved in the intact fusion constructs, eliminating the absolute necessity for
`intracellular release of the rGel component. Nevertheless, we explored a variety of
`different linker strategies to determine whether intentionally cleavable linkers
`offered an advantage over linkers which were designed for fl exibility only.
`Illustrations of various immunotoxin constructs are shown in Fig. 18.1a . The initial
`rGel-based immunotoxins consisted of a fl exible linker (GGGGS, “L”) tethering
`the C-terminus of the human anti-HER2/neu single-chain antibody (scFv) C6.5 to
`the native rGel N-terminus. The C6.5/rGel construct was further engineered by
`incorporating two different enzymatically sensitive furin cleavage linkers between
`
`IMMUNOGEN 2016, pg. 5
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`324
`
`Y. Cao and M.G. Rosenblum
`
`scFv and toxin components. The two furin-sensitive sequences designated “Fpe”
`(TRHRQPRGWEQL, 12 amino acid residues to PE273-284 sequence) and “Fdt”
`(AGNRVRRSVG, 10 amino acid residues to DT187-196 sequence), respectively.
`
` Tumor-Targeting scFv
`
` Previous studies identi fi ed a recombinant murine anti-HER2/neu scFv designated
`e23, and fusion constructs containing catalytic toxins such as PE and DT were
`shown to speci fi cally kill HER2/neu-expressing cells [ 33, 43 ] . A major drawback of
`such proteins is their potential for immune response after repeated administration.
`Repeated doses may cause hypersensitive reactions and lead to neutralization of the
`immunotoxins by antibodies directed against the nonhuman domains [ 77, 78 ] . The
`development of immunotoxins containing human or humanized components may
`circumvent these problems. Such immunotoxins may display reduced immunoge-
`nicity although antibodies to the toxin components may still limit prolonged therapy
` [ 79, 80 ] . We previously reported in vitro characterization and in vivo antitumor
`ef fi cacy studies of an immunotoxin composed of the human chimeric anti-HER2/
`neu antibody (BACH-250) chemically conjugated to rGel. The BACH-250/rGel
`conjugate demonstrated potent and speci fi c cytotoxicity against HER2/neu-overex-
`pressing human tumor cells in culture and against ovarian SKOV3 tumor xenografts
` [ 30 ] . However, the potential problems of diffusion of relatively large molecules
`such as full-length antibodies into solid tumors have been extensively addressed by
`Jain et al. [ 81 ] . The treatment of solid tumors presents a signi fi cant challenge since
`therapeutic antibodies must diffuse into the tumor against a hydrostatic pressure
`gradient and into disordered vasculature. These theoretical issues may not limit
`clinical response with antibodies alone since higher applied doses may circumvent
`these effects [ 81– 83 ] , but it may not be possible to overcome these limitations with
`higher doses of antibody–drug conjugates or immunotoxins because these agents
`frequently have a much narrower therapeutic window.
` Over the past decade, a variety of different anti-HER2/neu recombinant antibody
`formats have been engineered which are suitable for diverse therapeutic applica-
`tions and include monovalent, bivalent, and multivalent derivatives; single- or dou-
`ble-chain formats; and covalently or noncovalently linked assemblies of antibody
`heavy (V H ) and light chain variable domains (V L ) [ 84– 89 ] . Among these, human
`scFv appear to be effective when utilized as targeting domains incorporated into
`chimeric fusion proteins. They consist of antibody V H and V L sequences genetically
`linked via a fl exible linker, and they lack constant regions and Fc domains, thereby
`preventing possible binding to normal tissues and cells via interaction with Fc
`receptors. We previously engineered a series of novel fusion proteins created from
`various human anti-HER2/neu scFv designated C6.5 and various af fi nity mutants
`(designated ML3-9, MH3-B1, and B1D3, created by site-directed amino acid sub-
`stitutions in the CDR3s). The af fi nities of the scFvs ranged from 10 -8 to 10 -11 M
`(Fig. 18.1b ) [ 90, 91 ] . Recombinant immunotoxins containing each scFv and rGel
`were constructed by overlapping PCR and were designated C6.5/rGel, ML3-9/rGel,
`MH3-B1/rGel, and B1D3/rGel, respectively (Fig. 18.1c ).
`
`IMMUNOGEN 2016, pg. 6
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`18 Design, Development, and Characterization...
`
`325
`
` Functional Activity Analysis of Immunotoxins
`
` Tumor-antigen af fi nity and speci fi city of scFvs are important variables which may
`impact off-target tissue distribution and toxicity in vivo. These attributes have led to
`the commonly held concept that scFv must have high af fi nity in order to be thera-
`peutically relevant. However, studies by Adams et al. suggested that high-af fi nity
`scFv may be suboptimal vehicles and that lower-af fi nity scFv appear to diffuse more
`uniformly throughout the tumor interior [ 92, 93 ] . In addition, since the presence of
`shed tumor antigen has the potential to misdirect the targeted constructs through
`immune complex formation [ 64, 94 ] , higher-af fi nity scFv could potentially be less
`effective compared to lower-af fi nity constructs. Therefore, applying a series of
`rGel-containing fusion constructs composed of various linkers and scFv mutants
`with varying af fi nity to HER2/neu, we will examine the impact of construct design
`on in vitro cytotoxicity, pharmacodynamics, and antitumor ef fi cacy. Further inves-
`tigations included the effect of antibody af fi nity on behavior in the presence of sol-
`uble antigen, formation of immune complexes, and the coincident development of
`off-target toxicity.
`
` In Vitro Studies: Impact of Various Design Modi fi cations
`on In Vitro Immunotoxin Cytotoxic Activity
`
` Effects of Linker Design on Immunotoxin Potency and Functional Stability
`
` Based on C6.5/rGel containing the universal fl exible GGGGS linker, we introduced
`proteolytically cleavable linkers (Fpe and Fdt) and to examine whether this change
`would improve killing ef fi ciency. To investigate the susceptibility of various chime-
`ric toxins to proteolytic cleavage, puri fi ed fusions were subjected to proteolysis
`with recombinant furin. As indicated in Fig. 18.2a , the Fdt linker was the most sen-
`sitive to cleavage among all constructs tested. In contrast, cleavage of the molecule
`containing the Fpe linker was highly dependent on pH. As expected, the L linker
`was found to be comparatively resistant to intracellular protease action without
`regard to the pH.
` The intracellular release of rGel after endocytosis of various C6.5/rGel fusion
`constructs was next assessed in SKOV3 cells (Fig. 18.2b ). Although the maximal
`rGel release of different fusions was achieved at different time points, the absolute
`amounts of delivered rGel found in the cytosol were virtually identical. Therefore,
`this data con fi rms the observation that introduction of an unstable furin cleavage
`linker does not improve the intracellular rGel release of the constructs.
` The linkers tethering the C6.5 scFv and the rGel toxin demonstrated a differ-
`ential sensitivity to protease action which may result in different clearance and
`metabolic kinetics in vivo [ 95, 96 ] . We next performed a stability study of the
`constructs in human plasma (Fig. 18.2c, d ). The binding activity and cytotoxicity
`
`IMMUNOGEN 2016, pg. 7
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`a
`
`51-
`
`.. _
`
`~-----­
`
`ao-
`u-
`
`.. -.. ---
`b - 1 2 <t I U Jllh 1 2
`.. _
`•
`.. _
`25 - - -- ·---.. _ ~-- -
`
`.t I
`
`til<& Ill
`
`t J C I 11 Mil
`
`--
`
`-----------------
`
`
`~ - -
`t"N('lll
`
`-
`
`-
`
`11.1 • • • • 100 100
`
`c
`
`d
`
`60
`
`50
`
`~40
`c:
`~30
`!.2 20
`
`10
`
`0
`
`10
`
`60
`50
`40
`30
`20
`Incubation time (h)
`
`70
`
`80
`
`C6.5-l-tGel
`
`C6.5-l'pe-rGel
`
`C6 5-Fdl.fGel
`
`326
`
`
`
`Y. Cao and M.G. Rosenblum
`
` Fig. 18.2 Functional analysis of C6.5/rGel series immunotoxins in vitro. ( a ) Western blot analysis
`of furin cleavage of puri fi ed C6.5/rGel fusion constructs. ( b ) Western blot analysis of intracellular
`rGel release of C6.5/rGel fusions in SKOV3 cells. ( c , d ) 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 test
`
`of the C6.5–Fdt–rGel construct was shown to be the least stable after incubation
`because of the instability of the Fdt linker. On the other hand, of all molecules
`tested, the C6.5–L–rGel demonstrated the highest degree of stability after plasma
`incubation. The two companion molecules with the furin-cleavable linkers Fpe or
`Fdt demonstrated signi fi cantly less stability in vitro.
` Based on our studies, introduction of a furin-cleavable linker between C6.5 and
`rGel did not result in improved intracellular rGel release and cytotoxic effects
`in vitro, despite showing more sensitivity to protease cleavage and greater intracel-
`lular release of the rGel component. On the other hand, the rGel-based molecules
`with the furin-cleavable linkers demonstrated signi fi cantly less functional stability
`in vitro. The enzymatic stability of the linker can seriously affect the pharmacoki-
`netics of immunotoxins and can apparently impact loss of targeting function and
`in vivo ef fi cacy. Therefore, we clearly demonstrated the highly individualized nature
`of some payloads and targeted constructs and that observations regarding rGel may
`not necessarily translate to other payloads.
`
`IMMUNOGEN 2016, pg. 8
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`18 Design, Development, and Characterization...
`
`327
`
` The Impact of scFv Af fi nity on Immunotoxin Activity
`
` Although previous studies suggested that the binding af fi nity for antigen plays a
`pivotal role in the total concentration and penetration of scFv into tumors [ 92, 93 ] ,
`few companion studies have been conducted to determine whether scFv-based
`immunotoxins display the same behavior with regard to the relationship between
`af fi nity, tumor penetration, tumor residence, and ef fi cacy. Therefore, based on the
` fl exible GGGGS linker, we created a series of rGel-based immunotoxins from
`af fi nity mutants of the human anti-HER2/neu scFv C6.5.
` To ensure that immunotoxins retained antigen binding ability, the fusion proteins
`were compared in an ELISA-based binding assay using HER2/neu-positive (SKBR3,
`BT474 M1) and HER2/neu-negative (MCF7) cells. All the scFv/rGel constructs
`demonstrated speci fi c and signi fi cant ELISA binding to HER2/neu-positive cells
`with negligible binding to negative cells (Fig. 18.3a ). The equilibrium dissociation
`constants ( K d ) were calculated, and the af fi nities of immunotoxins for BT474 M1
`cells were found to be 53.13 nM (C6.5/rGel), 1.45 nM (ML3-9/rGel), 0.18 nM
`(MH3-B1/rGel), and 0.027 nM (B1D3/rGel). The correlation between the K d values
`of the scFvs and fusion constructs was found to be signi fi cant with a correlation
`coef fi cient of 0.939 ( p < 0.01), indicating that introduction of the rGel component
`did not affect the binding af fi nity of the scFv.
` We next examine the ability of various af fi nity scFv/rGel fusions to speci fi cally
`internalize into target cells. Immuno fl uorescence staining was performed on HER2/
`neu-positive and HER2/neu-negative cells after exposure to the constructs (Fig. 18.3b ).
`As quanti fi ed by relative fl uorescence (Fig. 18.3c ), the internalization ef fi ciency into
`HER2/neu-positive cells was shown to increase with increasing antibody af fi nity. For
`BT474 M1 cells, the relative fl uorescence intensities were 56.30 (C6.5/rGel), 73.69
`(ML3-9/rGel), 86.29 (MH3-B1/rGel), and 90.41 (B1D3/rGel). There was a good
`correlation between increases in apparent af fi nity and internalization ef fi ciency
`( r 2 = 0.8289; p < 0.01), indicating that ef fi cient binding to the cell surface appears to
`be primarily responsible for rapid internalization after cell exposure.
` The cytotoxicity of the various scFv/rGel constructs was then tested against a
`panel of different tumor cell lines (Table 18.2 ). As expected, there appeared to be a
`good correlation ( r 2 = 0.7812; p < 0.01) between apparent af fi nity and IC 50 values.
`The highest targeting indices were found for the highest-af fi nity construct (B1D3/
`rGel). Therefore, in vitro, binding af fi nity appeared to mediate internalization
`ef fi ciency, and this appeared to directly impact the overall cytotoxic effects observed.
`Furthermore, against HER2/neu-negative cells, there was little or no speci fi c cyto-
`toxicity of the constructs compared to rGel itself.
` Based on scFv/rGel immunotoxins, we demonstrated that increasing af fi nity
`could improve cell binding ability, internalization ef fi ciency, and cytotoxic activity
`on HER2/neu-positive cells. However, fusion toxins with a tenfold increase in
`af fi nity did not show a corresponding improvement in either internalization or a
`concomitant improvement in cytotoxic effects. This suggests that the internalization
`rate of the construct may primarily be associated with HER2/neu receptor recycling
`and the rate of antigen endocytosis and this may primarily be unaffected by the
`
`IMMUNOGEN 2016, pg. 9
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`~ 0.6 ""~ MCF7 (-)
`
`0.1
`o.
`0.6
`o.•
`0.3
`
`0 C6..,..
`....,.liiiGOI
`0
`A f*U.BllrGtl
`. .....
`• at~
`
`0.1
`
`3 0,5
`~ 0 ,4
`; 0.3
`~ 0.2
`~ 0.1
`0.0
`0.01
`
`0.1
`
`10
`
`100
`
`cone. nt.t
`
`0.1
`
`10
`
`100
`
`Cone. nt.t
`
`0.2
`
`0.1
`o.o
`0.01
`
`0.1
`
`10
`
`100
`
`cone. nM
`
`C6.5/rGel
`
`Ml3·9/rGel
`
`MH3·Bl/rGel
`
`8103/rGel
`
`rGel
`
`b
`
`SKBR3
`
`BT474 Ml
`
`MCF7
`
`a
`0,1
`o.•
`0.6
`o.•
`0.3
`
`0.2
`
`0.1
`
`0.0
`0 .0 1
`
`c
`
`328
`
`Y. Cao and M.G. Rosenblum
`
` Fig. 18.3 Characterization and comparison of different scFv/rGel immunotoxins. ( a ) Evaluation
`binding activity of the fusion constructs to HER2/neu-positive (SKBR3 and BT474 M1) and HER2/
`neu-negative (MCF7) cells by whole-cell ELISA. ( b ) Internalization of the immunotoxins on HER2/
`neu-positive and HER2/neu-negative cells. Cells were subjected to immuno fl uorescent staining
`with anti-rGel antibody (FITC-conjugated secondary), with propidium iodine nuclear counterstain-
`ing. ( c ) Quanti fi cation of internalization rate of the immunotoxins. The bar graphs were calculated
`from relative fl uorescence estimation, and the values are expressed as mean ± S.D ( n > 50)
`
`af fi nity of immunotoxin binding [ 97, 98 ] . Moreover, intracellular traf fi cking and
`distribution of the toxin component to the ribosomal compartment may be addi-
`tional critical factors which can de fi ne immunotoxin sensitivity [ 99 ] .
`
` Mechanistic Studies of Immunotoxin Cytotoxic Activity
`
` Studies of bacterial and plant immunotoxins have provided us with essential infor-
`mation regarding intracellular routing and translocation events [ 100, 101 ] , and this
`
`IMMUNOGEN 2016, pg. 10
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`18 Design, Development, and Characterization...
`
`329
`
` Table 18.2 Comparative cytotoxicity of rGel-based fusion constructs against various tumor cell
`lines
`
` IC 50 (nM) (TI) *
` HER2/neu
` MH3-B1/
` ML3-9/
` Tumor
`Expression
`rGel
`rGel
` C6.5/rGel
`type
` Cell line
`level
` 2.7 (387)
` 5.0 (211)
` 6.4 (165)
` ****
` Breast
` SKBR3
` 3.9 (56)
` 10.9 (20)
` 18.9 (12)
` ****
` Breast
` BT474 M1
` 9.1 (106)
` 20.4 (47)
` 30.1 (32)
` ****
` Gastric
` NCI-N87
` 13.0 (41)
` 18.1 (29)
` 24.3 (22)
` ****
` Lung
` Calu3
` 155 (2)
` 149.9 (2)
` 145.8 (2)
` *
` Breast
` MDA MB231
` 260.6 (1)
` 246.7 (1)
` 246.9 (1)
` *
` Breast
` MCF7
` 153.9 (1)
` 126.9 (2)
` 61.4 (3)
` *
` Melanoma
` A375m
` 194.6 (1)
` 185.4 (1)
` 160.8 (1)
` *
` Cervical
` Me180
` *Targeting index (TI) represents IC 50 of rGel/IC 50 of immunotoxin
`
` B1D3/rGel
` 1.9 (567)
` 1.2 (177)
` 4.9 199)
` 10.0 (53)
` 204.8 (1)
` 266.9 (1)
` 173.9 (1)
` 213.1 (1)
`
` rGel
` 1061
` 219.8
` 965.6
` 531.1
` 297.3
` 247.4
` 207.3
` 222.5
`
`information can be employed in the design of optimized constructs. The mechanism
`of cytotoxicity of anti-HER2/neu immunotoxins generally involves inhibition of
`cellular protein synthesis, although the point of attack in this pathway can vary
`slightly depending on the toxin [ 102, 103 ] .
` The plant-derived toxins are primarily ribosome-inactivating proteins (RIPs) and
`are enzymes which depurinate rRNA, thus inhibiting protein synthesis. They may
`also depurinate other polynucleotide substrates [ 104– 106 ] . rGel is an N -glycosidase
`generating cytotoxic effects which are the direct result of protein synthesis inhibi-
`tion (Fig. 18.4a ). Although there have been numerous preclinical studies of rGel-
`based immunotoxins for the treatment of both solid and hematological tumors, the
`actual mechanisms behind the induction of cell death appear to vary depending on
`the cell type targeted [ 107– 109 ] . This occurs because the basic mechanism of pro-
`tein synthesis inhibition due to the rGel component causes the loss of different high-
`turnover proteins in cells derived from different tumor types. This fi nally results in
`cytotoxic patterns and mechanisms which can vary widely based on the proteins
`critical for survival of different cell types targeted.
` The cytotoxic effects mediated by our rGel-based immunotoxins were analyzed
`including apoptosis, necrosis, and autophagy in BT474 M1 cells. As shown in
`Fig. 18.4b , scFv/rGel fusions did not activate caspase-dependent apoptosis in tar-
`get cells, showing no cleavage of the caspase substrate PARP. We next assessed
`lactate dehydrogenase (LDH) release, a marker of abrupt membrane lysis, and
`found that exposure of BT474 M1 cells to immunotoxins did not induce necrotic
`cell death using this parameter (Fig. 18.4c ).
` We next examined whether the cytotoxic effects of these immunotoxins activate
`autophagic signaling. As shown in Fig. 18.4d , the ratio of LC3-II formation to the
` b -actin control was shown to be increased after treatment with the fusion constructs,
`demonstrating that autophagic fl ux was induced by rGel-based immunotoxins in
`BT474 M1 cells. In addition, autophagic induction by fusion constructs was further
`validated by the observed selective release of cellular high mobility group box 1
`
`IMMUNOGEN 2016, pg. 11
`Phigenix v. Immunogen
`IPR2014-0676
`
`

`
`a
`
`Tumor cells
`
`·····-·······
`
`Herl/neu homodimers
`Or heterodimers
`
`Turnover Proteins
`
`~~
`·- - __..,..
`~ Inhibition of
`Protein synthesis
`c
`
`0
`
`e
`_;////
`
`...,
`- ..
`- ;///)'
`- a •
`-------
`
`LC4 ~~-~ HMG81
`Lc•
`,_,.,. -
`
`C.lla
`
`Medium
`
`-t~/h-7 - t~//,{/
`,_., -
`
`--
`
`b
`
`14h
`
`d
`Nil
`
`LC•
`LC•
`
`.......
`
`330
`
`
`
`Y. Cao and M.G. Rosenblum
`
` Fig. 18.4 Cell-killing mechanism analysis of the immunotoxins on BT474 M1 cells. ( a ) A gener-
`alized view of cytotoxic mechanism of anti-HER2/neu scFv/rGel fusions in targeted cells. ( b )
`Western blot analysis of PARP cleavage after 24 and 48 h of scFv/rGel fusions treatment. ( c )
`Evaluation of necrosis by LDH release after treatment with fusion proteins or Triton X-100
`(mean ± S.D. from three replicates). ( d ) Analysis of LC3 after treated with scFv/rGel fusions for 24
`and 48h. ( e ) Analysis of cell extract and medium for HMGB1 protein after immunotoxins treat-
`ment for 48 h
`
`(HMGB1) (Fig. 18.4e ). Tumor cells undergoing autophagy can s

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