throbber
Protein Engineering vol.13 no.8 pp.565–574, 2000
`
`ScFv multimers of the anti-neuraminidase antibody NC10:
`shortening of the linker in single-chain Fv fragment assembled in
`VL to VH orientation drives the formation of dimers, trimers,
`tetramers and higher molecular mass multimers
`
`Olan Dolezal1, Lesley A.Pearce, Lynne J.Lawrence2,
`Airlie J.McCoy3, Peter J.Hudson and Alexander A.Kortt
`
`CSIRO Health Sciences and Nutrition and CRC for Diagnostic
`Technologies, 343 Royal Parade, Parkville, Victoria, 2Biomolecular
`Research Institute, 343 Royal Parade, Parkville, Victoria, Australia 3052 and
`3Department of Haematology, Cambridge University, Hills Road, Cambridge
`CB2 2XY, UK
`1To whom correspondence should be addressed.
`E-mail: olan.dolezal@hsn.csiro.au
`Synthetic genes encoding single-chain variable fragments
`(scFvs) of NC10 anti-neuraminidase antibody were con-
`structed by joining the VL and VH domains with linkers of
`fifteen, five, four, three, two, one and zero residues. These
`VL–VH constructs were expressed in Escherichia coli and
`the resulting proteins were characterized and compared
`with the previously characterized NC10 scFv proteins
`assembled in VH–VL orientation. Size-exclusion chromato-
`graphy and electron microscope images of complexes
`formed between various NC10 scFvs and anti-idiotype Fab⬘
`were used to analyse the oligomeric status of these scFvs.
`The result showed that as the linker length between VL
`and VH was reduced, different patterns of oligomerization
`were observed compared with those with VH–VL isomers.
`As was the case for VH–VL orientation, the scFv-15 VL–
`VH protein existed mainly as a monomer whereas dimer
`(diabody) was a predominant conformation for the scFv-5,
`scFv-4 and scFv-3 VL–VH proteins. In contrast to the VH–
`VL isomer, direct ligation of VL to VH led to the formation
`of predominantly a tetramer (tetrabody) rather than to an
`expected trimer (triabody). Furthermore, the transition
`between dimers and higher order oligomers was not as
`distinct as for VH–VL. Thus reducing the linker length in
`VL–VH from three to two residues did not precisely dictate
`a transition between dimers and tetramers. Instead, two-
`residue as well as one-residue linked scFvs formed a
`mixture of dimers, trimers and tetramers.
`Keywords: antibody/diabody/single-chain Fvs/tetrabody/tria-
`body
`
`Introduction
`The antigen-binding portion of an antibody molecule (Fv
`fragment) is formed by the association of the heavy chain
`variable region (VH) and the light chain variable region (VL).
`Fv fragments are the smallest entities that consistently maintain
`the binding specificity of the whole antibody. Although recom-
`binant DNA techniques have facilitated individual Fv domain
`production, the non-covalently associated VL and VH domains
`in an individual Fv fragment tend to dissociate from one
`another (Glockshuber et al., 1990). To improve stability,
`recombinant single chain Fv fragments (scFvs) have been
`engineered with two variable domains covalently joined via a
`flexible peptide linker (Bird et al., 1988; Huston et al., 1988).
`In general, scFvs with linkers ⬎12 amino acid residues in
`
`© Oxford University Press
`
`length form stable monomers, which usually exhibit similar
`antigen binding affinity to the parent antibody (Skerra et al.,
`1991; Kortt et al., 1994). However, scFv monomers have also
`been observed to form active dimers and higher molecular
`mass multimers upon freezing at high protein concentrations
`(Kortt et al., 1994).
`To provide increased avidity to target antigens, recent
`attention has focused on the design of linkers which generate
`multivalent scFv molecules (reviewed by Hudson, 1999).
`Multivalent scFv molecules are sufficiently large to avoid the
`fast clearance through the kidneys observed for scFv monomers
`and thereby have potential application for tumour imaging and
`radiotherapy (Adams et al., 1998; Colcher et al., 1998; Wu
`et al., 1999). Construction of stable multimeric scFv molecules
`can be achieved by the shortening of the linker length to ⬍12
`residues such that the VH domain is unable to associate with
`its attached VL domain and thus generate a monomeric Fv
`fragment. Instead, VH and VL domains from one scFv molecule
`associate with VH and VL domains from a second scFv
`molecule to form a bivalent dimer, termed a diabody (Hollinger
`et al., 1993). When the linker is shortened to ⬍3 residues or
`when VH and VL domains are directly ligated to each other,
`scFv molecules associate to form a trimer, termed a triabody
`(Iliades et al., 1997; Kortt et al., 1997).
`ScFvs of both VH–linker–VL and VL–linker–VH orientation
`have been produced by various research groups (Huston et al.,
`1991). Our laboratory has to date almost exclusively produced
`scFvs with the VH domain at the amino terminus (Malby et al.,
`1993, Lilley et al., 1994; Coia et al., 1997; Iliades et al.,
`1997). In particular, NC10 anti-neuraminidase scFv antibody,
`in a VH–linker–VL orientation, has been used extensively to
`elucidate the oligomeric nature of scFvs with reduced linker
`lengths (Kortt et al., 1997; Atwell et al., 1999). To investigate
`the oligomerization phenomenon further, the NC10 scFv frag-
`ment was assembled in a reverse, VL–linker–VH, orientation.
`The observed result showed that as the linker length between
`VL and VH was reduced, different patterns of oligomerization
`were observed compared with the original VH–linker–VL
`orientation. Furthermore, the direct ligation of VL to VH led
`to the formation of a tetramer, rather than to a trimer as
`observed for direct ligation of VH to VL.
`
`Materials and methods
`Sequence numbering
`Antibody residues were numbered according to Kabat et al.
`(1991) and for NC10 correspond exactly to Malby et al. (1993).
`Residues in the VL domain of the scFv were superscripted with
`L and the residue number; for example, ArgL107 signifies
`arginine in position 107 of the VL domain. Similarly residues
`in the VH domain of the scFv were superscripted H and the
`residue number.
`General cloning procedures
`Unless stated otherwise, all DNA manipulations were carried
`out according to standard protocols (Sambrook et al., 1989)
`
`565
`
`UPenn Ex. 2062
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`O.Dolezal et al.
`
`with reagents purchased from New England Biolabs. All
`polymerase chain reactions (PCRs) were performed with Pfu
`DNA polymerase (Stratagene). The PCR-amplified DNA frag-
`ments were digested with appropriate restriction enzymes, run
`on 1% (w/v) agarose gel and purified from the gel using
`BRESAclean purification kit (Bresatech). The purified DNA
`fragments were ligated into similarly prepared expression
`vectors using reagents and protocols supplied by Gibco-BRL.
`The ligation mixtures were transformed by the electroporation
`method (Dower et al., 1988) into Escherichia coli XL 1-Blue
`MRF⬘ cells (Stratagene) and recombinant clones were identified
`by colony PCR using primers complementary to 5⬘ and 3⬘
`ends of recombinant gene inserts. All DNA sequences of
`various scFv constructs were verified using Dye Terminator
`Cycle Sequencing kits with AmpliTaq (PE Applied Bio-
`systems).
`Construction of NC10 scFv gene fragments with 0 and 15
`residue linkers
`The previously described pPOW–NC10 scFv-15 (VH–VL) gene
`construct (Malby et al., 1993) was used as a source of VL and
`VH gene fragments. To generate the NC10 scFv-0 (VL–VH)
`synthetic gene, the VL and VH gene fragments were PCR-
`amplified using primers N4311 and N4341 for VL, and N4342
`and N4293 for VH (Table I). The resulting VL and VH PCR
`products were gel-purified and joined into an scFv-0 using
`PCR overlap extension (Horton et al., 1989). To create the
`NC10 scFv-15 (VL–VH) synthetic gene, VL and VH gene
`fragments were PCR-amplified using primers N4311 and
`N4535 for VL, and N4534 and N4293 for VH (Table I). The
`resulting VL and VH gene fragments each contained part of
`the linker sequence at the 5⬘ and 3⬘ ends, respectively, as well
`as the BamHI restriction site which allowed for correct in
`frame ligation of these V fragments via a linker sequence
`encoding 15 amino acid (GGGGS)3. The NC10 scFv-0 and
`scFv-15 (VL–VH) synthetic genes were then digested with
`NcoI and NotI restriction enzymes and cloned separately into
`a likewise digested pGC-4C2 E.coli vector (Coia et al., 1997)
`to create pGC-NC10 scFv-0 and scFv-15 plasmids (Figure 1a-
`i). This pGC-4C2 vector backbone incorporates two, rather
`than one, C-terminal FLAG peptide epitopes (Hopp et al.,
`1988) for improved purification efficacy of FLAG-fusion
`proteins. The resulting NC10 scFv-0 and scFv-15 gene con-
`structs encoded N-terminal cleavable pelB periplasmic target-
`ing signal followed by NC10 scFv and two C-terminal FLAG
`peptide epitope tags which were linked to VH domain at the
`C-terminus by three alanine residues and separated from one
`another by two alanine residues (Figure 1a-iii). The NC10
`scFv-15 and scFv-0 (VL–VH) gene fragments were also inserted
`into a heat-inducible pPOW vector (Power et al., 1992). This
`was done by digesting the above described pGC-NC10 scFv-
`15 and scFv-0 (VL–VH) plasmids with NcoI and EcoRI
`restriction enzymes and ligating the scFv synthetic genes into
`a likewise digested pPOW vector. Similarly as for pGC
`constructs,
`the resulting gene fragment (Figure 1a-ii) was
`designed to express pelB signal sequence followed by NC10
`scFv and two C-terminal FLAG tags.
`Construction and cloning of NC10 scFv gene fragments with
`shorter linkers
`The pGC-NC10 scFv-0 (VL–VH) gene construct (Figure 1a)
`was used for insertion of linkers of increasing length. This
`vector was digested simultaneously with XhoI and PstI restric-
`tion enzymes and the remaining plasmid DNA was purified
`
`566
`
`Fig. 1. (a) Schematic diagram of the NC10 scFv (VL–VH) expression unit
`in pGC (i) and pPOW (ii) vectors. (iii) Important parts of nucleotide and
`amino acid sequences of the NC10 scFv (VL–VH) unit in pGC and pPOW,
`outlining restriction sites for cloning (underlined), C-terminal region of pelB
`sequence, N-terminal sequence of VL gene and C-terminal sequence of VH
`gene terminal (in bold), and two C-terminal octapeptide FLAG epitopes (in
`bold). (b) Amino acid sequences of the C-terminus of the VL domain,
`N-terminus of the VH domain and of the linker peptide (in bold) used in
`each of the NC10 scFv constructs.
`
`using agarose gel. Five sets of synthetic oligonucleotides
`(Table I) were phosphorylated at the 5⬘ termini by incubation
`at 37°C for 30 min with 0.5 units of T4 polynucleotide kinase
`and 1 mM rATP (Biotech International) in 1⫻ PNK buffer.
`Pairs of complementary phosphorylated oligonucleotides were
`pre-mixed in equimolar ratios to form DNA duplexes that
`encoded linkers of increased length. The one-residue linker
`construct was designed to encode a Ser residue in between
`codons for C-terminal VL ArgL107 and N-terminal VH GlnH1,
`whereas the two-, three-, four- and five-residue linker constructs
`were designed to encode additional glycine residues immedi-
`ately preceding the Ser linker residue (Figure 1b). All five of
`these duplexes were designed to contain a ‘sticky end’ overlap
`compatible with XhoI and PstI restriction enzyme sites at the
`5⬘ and 3⬘ ends, respectively. This allowed for direct cloning
`of these duplexes into XhoI/PstI restricted pGC-NC10 scFv-0
`(VL/VH) plasmid.
`
`UPenn Ex. 2062
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`Table I. Synthetic oligonucleotides used for construction of NC10 scFv VL–VH gene fragments
`
`scFv multimers NC10scFVmultimers
`
`TCG AGA TAA GAG GTG GAG GTG GAT CCC AGG TGC AGC TGC A
`GCT GCA CCT GGG ATC CAC CTC CAC CTC TTA TC
`
`TCG AGA TAA GAG GAG GTG GAT CCC AGG TGC AGC TGC A
`GCT GCA CCT GGG ATC CAC CTC CTC TTA TC
`
`TCG AGA TAA GAG GTG GAT CCC AGG TGC AGC TGC A
`GCT GCA CCT GGG ATC CAC CTC TTA TC
`
`scFv–15 and scFv–0
`N4311:
`AAA ACC ATG GCC GAT ATC GAG CTC ACA CAG
`N4341:
`AGA CTG CTG CAG CTG CAC CTG TCT TAT CTC GAG CTT GGT
`N4342:
`GGG ACC AAG CTC GAG ATA AGA CAG GTG CAG CTG CAG CAG TCT
`N4293:
`TTT TGC GGC CGC GGA GAC GGT GAC CGT GGT CCC
`N4535:
`TTT GGA TCC ACC TCC ACC GCA ACC GCC TCC ACC TCT TAT CTC GAG CTT GGT CCC
`N4534:
`AAA GGA TCC GGT GGA GGC GGT TGC CAG GTG CAG CTG CAG CAG
`scFv–5
`N5006:
`N5009:
`scFv–4
`N5007:
`N5010:
`scFv–3
`N5008:
`N5004:
`scFv–2
`N5011:
`N5003
`scFv–1
`N5005
`N5002
`
`TCG AGA TAA GAG GAT CCC AGG TGC AGC TGC A
`GCT GCA CCT GGG ATC CTC TTA TC
`
`TCG AGA TAA GAT CCC AGG TGC AGC TGC A
`GCT GCA CCT GGG ATC TTA TC
`
`Oligonucleotide sequences are listed 5⬘ to 3⬘. Linker- and vector-related gene sequences are in italics.
`Important restriction sites are underlined.
`
`Expression and purification of the scFvs
`NC10 scFv-15 and scFv-0 gene constructs (VL–VH) were
`initially expressed in a heat-inducible pPOW vector (Power
`et al., 1992). Each pPOW-NC10 scFv construct was expressed
`in 1 l of 2YT/amp100 as described by Malby et al. (1993),
`using E.coli strain TOP 10F⬘ (Invitrogen) as host. The scFv-15
`and scFv-0 proteins (VL–VH) were located in the periplasm as
`insoluble aggregates associated with the membrane fraction,
`as found previously for NC10 scFv VH–VL proteins (Malby
`et al., 1993; Kortt et al., 1997). Recombinant scFv proteins
`were purified using a modified protocol of Kortt et al. (1997).
`Cells were disrupted by sonication in 100 ml of phosphate-
`buffered saline, pH 7.4 (PBS), centrifuged and the insoluble
`aggregates of NC10 scFvs were solubilized in 100 ml of 6 M
`guanidine hydrochloride, 0.1 M Tris–HCl, pH 8.0. The resulting
`mixture was then dialysed against PBS and insoluble material
`removed by centrifugation. Recombinant scFvs were purified
`from soluble fractions (supernatant) using an M2 anti-FLAG
`IgG-Sepharose column (5⫻1 cm; Brizzard et al., 1994). The
`affinity column was equilibrated in PBS and bound proteins
`were eluted with 100 mM glycine, pH 3.0. The eluted proteins
`were neutralized with 1/10 volume of 1 M Tris–HCl (pH 8.0)
`and dialysed extensively against PBS–0.02% (w/v) sodium
`azide. Proteins samples were concentrated by ultrafiltration
`(Amicon) over a 10 kDa cut-off membrane (YM10, Diaflo) to
`~1 mg/ml and stored at 4°C.
`Expression and purification of NC10 scFvs (VL–VH) with
`shorter linkers
`Each pGC-NC10 scFv (VL–VH) construct was expressed in
`500 ml of 2YT/amp100 ⫹ 0.1% (w/v) D-glucose as described
`in Dolezal et al. (1995), using E.coli strain TOP 10F⬘ cells as
`host. Expression experiments were terminated 3 h post-induc-
`tion and proteins were isolated from the periplasmic space
`using a modified method of Minsky et al. (1986). Briefly, cells
`were centrifuged at 5000 g for 10 min and supernatant was
`discarded. After washing the cell pellet in ice-cold PBS, cells
`
`were resuspended in 20 ml of ice-cold spheroplast buffer (100
`mM Tris–HCl, pH 8.0, 0.5 M sucrose, 2 mM EDTA, 100 µg/
`ml lysozyme). After incubating the cell mixture on ice for 20
`min, ice-cold half-strength spheroplast buffer (50 mM Tris–
`HCl, pH 8.0, 1 mM EDTA, 0.25 M sucrose) was added to a
`final volume of 100 ml. Cells were mixed gently and left on
`ice for a further 30 min. Periplasmic proteins were harvested
`by centrifugation at 10 000 g. The supernatant (100 ml) was
`sonicated briefly to shear the remaining DNA/RNA and filtered
`through 0.45 µm filter. An Amicon concentrator unit (with
`YM10 membrane) was used to concentrate the periplasmic
`fraction to ~15 ml. Recombinant NC10 scFvs (VL–VH) proteins
`were affinity purified from the periplasmic fraction using an
`M2 anti-FLAG IgG-Sepharose column as described in the
`previous section. Eluted scFvs proteins were dialysed against
`PBS–0.02% (w/v) sodium azide, concentrated by ultrafiltration
`(YM10, Diaflo) to ~1 mg/ml and stored at 4°C.
`Biochemical and biophysical characterization of NC10 scFv
`proteins
`The purity of the NC10 scFvs was monitored by SDS–PAGE
`and Western blot analysis as described previously (Kortt
`et al., 1994). The concentrations of the scFv fragments were
`determined spectrophotometrically, using values for the extinc-
`tion coefficient (ε0.1%) at 280 nm of 1.66 for VL–VH scFvs
`and 1.70 for VH–VL scFvs calculated as described by Gill and
`von Hippel (1989). The relative molecular mass of each
`affinity-purified NC10 scFv was estimated using size-exclusion
`chromatographic columns (Superose 12 HR10/30 and/or Super-
`dex 200 HR 10/30, Pharmacia) on an HPLC system (Bio-Rad
`Model 700) at 21°C in PBS which were previously calibrated
`with Bio-Rad Gel Filtration Standard proteins. The flow-rate
`was 0.5 ml/min, and the absorbance of the effluent stream was
`monitored at both 214 and 280 nm. Elution times of various
`VL–VH scFv oligomers were compared with those already
`established for VH–VL scFv monomers, dimers and trimers
`(Kortt et al., 1994, 1997; Atwell et al., 1999).
`
`567
`
`UPenn Ex. 2062
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`O.Dolezal et al.
`
`Formation of complexes with 3–2G12 anti idiotype Fab⬘
`Fab⬘ fragments of the NC10 anti-idiotype antibody, 3–2G12,
`were prepared as described in Kortt et al. (1997). Purified
`NC10 scFv-0, scFv-1, scFv-2 and scFv-5 (VL–VH) proteins
`were mixed with a small molar excess of 3–2G12 anti-idiotype
`Fab⬘, as described previously by Kortt et al. (1997). The
`complexes were separated from excess Fab⬘ by size-exclusion
`chromatography on Superdex 200 (HR 10/30) in PBS, pH 7.4,
`with a flow-rate of 0.5 ml/min. The column had previously
`been calibrated with uncomplexed scFvs and 3–2G12 Fab⬘.
`Electron microscopy
`Complexes of scFv-0 and scFv-1, scFv-2 and scFv-5 with 3–
`2G12 Fab⬘ and the complex of influenza virus neuraminidase
`(soluble tetrameric extracellular domain) with NC10 Fab⬘
`(Malby et al., 1994) were examined by electron microscopy
`(EM). EM imaging and data analysis were performed as
`described previously (Lawrence et al., 1998; Atwell et al.,
`1999).
`Molecular modeling
`Computer-generated models of NC10 scFv triabodies and
`tetrabodies were constructed using Fv modules that corre-
`sponded to the coordinates of the NC10 Fv domain in PDB
`entry 1NMB (Malby et al., 1994, 1998). Fv modules were
`manipulated as rigid bodies with the O molecular graphics
`package (Jones et al., 1991). Triabody structure corresponded
`to the model described by Kortt et al. (1997) comprising three
`Fv modules with threefold symmetry. Tetrabodies comprised
`four Fv modules with fourfold symmetry. No attempt was
`made to model conformational changes in the Fv domains.
`
`Results and discussion
`Expression of NC10 scFv-15 and scFv-0 constructs
`For direct comparison with the original NC10 scFv-15 and
`scFv-0 (VH–VL) constructs (Malby et al., 1993; Kortt et al.,
`1997), the NC10 scFv-15 and scFv-0 (VL–VH) gene fragments
`were initially constructed and expressed in a heat-inducible
`expression vector pPOW (Figure 1a-iii). As for VH–VL orienta-
`tion, in the VL–VH construct of the NC10 scFv, the VL domain
`was linked to the VH domain using the classical linker design
`of Huston et al. (1988, 1991), whereby the codon for C-
`terminal VL ArgL107 was linked to the codon for N-terminal
`VH GlnH1 via a 15 amino acid residue linker ([G4S]3; Figure
`1b). The ArgL107 was defined from the NC10 scFv crystal
`structure to be the last residue in the VL that made intra-
`domain contacts within VL and additional residues therefore
`formed a true linker (Malby et al., 1998). In case of scFv-0,
`the C-terminal VL ArgL107 was ligated directly to VH GlnH1
`(Figure 1b).
`The pPOW/NC10 scFv-15 and scFv-0 (VL–VH) constructs
`were expressed under the same conditions as those used
`previously for the VH–VL constructs (Malby et al., 1993; Kortt
`et al., 1997). As was the case for the VH–VL proteins, the
`majority of expressed scFv-15 and scFv-0 (VL–VH) proteins
`were located in the periplasm as insoluble protein aggregates
`which were solubilized by extraction with 6 M GuHCl and
`on dialysis refolded into soluble, functional scFv entities.
`Furthermore, there was no apparent difference in expression
`levels between the VL–VH and VH–VL scFvs proteins. The
`resulting soluble scFvs were purified by affinity chromato-
`graphy on an M2 anti-FLAG IgG-Sepharose column. Yields
`of 2–5 and 1–2 mg of soluble affinity-purified protein per litre
`
`568
`
`Fig. 2. Size-exclusion chromatography on a calibrated Superose 12 HR 10/
`30 column of affinity-purified NC10 scFv-15 proteins. Superimposed on the
`same scale are runs for scFv-15 VL–VH (solid line) and scFv-15 VH–VL
`(dashed line). The VL–VH protein eluted as monomer at 28.0 min and dimer
`at 25.5 min. These elution times are consistent with calculated molecular
`masses of 28.5 and 57 kDa, respectively. The VH–VL protein eluted as
`monomer at 28.6 min and dimer at 26.2 min. These elution times are
`consistent with calculated molecular masses of 27 and 54 kDa, respectively.
`The column was equilibrated in PBS, pH 7.4, and the flow-rate was 0.5 ml/
`min.
`
`of shake-flask culture were typically obtained for scFv-15 and
`scFv-0, respectively.
`Biophysical analysis of NC10 scFv-15 and scFv-0 proteins
`Samples of affinity-purified scFv-15 proteins (VL–VH and
`VH–VL) were shown by SDS–PAGE to comprise essentially
`homogeneous scFv preparations with a main protein band at
`~27.0 kDa for VH–VL and ~28.5 kDa for VL–VH (data not
`shown). This apparent difference in protein mobility on SDS–
`PAGE was attributed to the additional sequence associated
`with the second FLAG epitope that has been added to the C-
`terminus of scFv VL–VH proteins (see Materials and methods
`and Figure 1a). Size-exclusion chromatography on a calibrated
`Superose 12 column showed that the oligomeric status of the
`NC10 VH–VL and VL–VH scFv-15 proteins was similar in
`solution (Figure 2). Gel filtration of affinity-purified scFv-15
`VL–VH protein showed the presence of two main peaks with
`apparent molecular masses of ~28.5 kDa (major peak) and
`~57.0 kDa (minor peak), corresponding to scFv-15 monomer
`and dimer, respectively. The scFv-15 VH–VL protein when
`analysed on the same column eluted mainly as monomer at
`~27.0 kDa with a small amount of dimer at ~54.0 kDa. The
`relative differences in elution times for VL–VH and VH–VL
`monomers and dimers were again attributed to the additional
`FLAG sequence at the C-terminus of scFv-15 VL–VH protein.
`Both scFv-15 protein samples also contained traces of higher
`order oligomers, but because of small quantities of these
`species it was not possible to assign their oligomeric status
`unequivocally. Similarly as shown for NC10 scFv-15 VH–VL
`protein (Kortt et al., 1994), the formation of NC10 scFv-15
`VL–VH dimers and higher molecular mass multimers was
`induced by storing the sample at higher concentrations (⬎1
`mg/ml) or by repeated freezing and thawing (data not shown).
`In contrast to scFv-15 proteins, size-exclusion chromato-
`graphy of scFv-0 (VL–VH and VH–VL) proteins demonstrated
`a significant difference in elution profiles (Figure 3a). The
`chromatography of the NC10 scFv-0 (VL–VH) protein yielded
`a major protein peak eluting at ~21.9 min with a distinct
`
`UPenn Ex. 2062
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`scFv multimers NC10scFVmultimers
`
`Fig. 4. Reducing SDS–PAGE of affinity-purified NC10 scFvs (VL–VH)
`stained with Coomassie Brilliant Blue G-250. Lanes: 1, scFv-0; 2, scFv-1;
`3, scFv-2; 4, scFv-3; 5, scFv-4; 6, scFv-5; 7, scFv-15.
`
`vector because of its capacity to produce soluble and active
`scFv proteins in the bacterial periplasm. A schematic diagram
`depicting the general outline of these short-linkered NC10
`scFv expression units in pGC is shown in Figure 1b. The pGC
`NC10 scFv VL–VH constructs were expressed in E.coli TOP
`10F⬘ cells using the expression protocol of Dolezal et al.
`(1995). The expressed scFv products were isolated from the
`periplasmic fraction by affinity chromatography as described
`in Materials and methods. The yield of soluble affinity-purified
`protein decreased progressively as the linker length was
`shortened,
`from 2 mg/l bacterial culture for scFv-15 to
`0.5 mg/l bacterial culture for scFv-0.
`Molecular mass analysis of scFv proteins with shorter
`linkers
`SDS–PAGE analysis of affinity-purified NC10 scFv-0, scFv-1,
`scFv-2, scFv-3, scFv-4, scFv-5 and scFv-15 (VL–VH) protein
`samples showed that the scFvs comprised predominantly a
`single component of apparent molecular mass ~27.5–28.5 kDa,
`as expected for this series of proteins (Figure 4). When
`these scFvs were subjected to analysis by size-exclusion
`chromatography on a calibrated Superdex 200 column, a
`number of major protein peaks were observed with a significant
`variation in elution times consistent with the presence of scFv
`oligomers (Figure 5). Elution profiles for scFv-5, scFv-4 and
`scFv-3 (Figure 5a) showed the presence of a single major peak
`with an elution time consistent with a molecular mass of ~57
`kDa expected for a dimer. This peak eluted at the same time
`as the dimer peak in scFv-15 (VL–VH) preparations (Figure
`5b). The minor higher molecular mass species observed in
`these three profiles (Figure 5a) eluted at the same elution times
`as the zero-linked NC10 VL–VH trimer and tetramer peaks
`(Figure 5b). These findings are consistent with those observed
`for the NC10 scFv-5, scFv-4 and scFv-3 (VH–VL) proteins
`which mainly formed dimer and a small amount of trimer
`(Atwell et al., 1999). However, no tetramer species was
`observed for scFv-5, -4 and -3 (VH–VL) proteins. Interestingly,
`in the case of scFv-5 (VL–VH) protein, consistently larger
`yields of tetramer species were observed and this allowed for
`the purification and subsequent characterization of this tetramer
`by gel filtration on a Superdex 200 column and by electron
`microscopy (see below). This scFv-5 tetramer was, however,
`relatively unstable as it partially reverted back to dimer (40%
`dimer after 24 h at 4°C). Similarly, a small amount of purified
`scFv-5 dimer species converted to tetramer (~5–10% after
`24 h at 4°C).
`
`569
`
`Fig. 3. Size-exclusion chromatography on a calibrated Superdex 200 HR 10/
`30 column of affinity-purified NC10 scFv-0 proteins. Columns were
`equilibrated in PBS, pH 7.4, and the flow-rate was 0.5 ml/min. (a) Shows
`the scFv-0 VL–VH tetramer (solid line) eluting at 21.9 min with a trimer
`shoulder on the trailing edge. Superimposed on the same scale is a run for
`scFv-0 VH–VL trimer (dashed line) eluting at 24.1 min. (b) Shows
`separation of scFv-0 tetramer from scFv-0 trimer using two Superdex 200
`HR 10/30 columns linked in tandem.
`
`shoulder on the trailing edge of the peak. The NC10 scFv-0
`(VH–VL) protein, previously demonstrated to be a trimer (Kortt
`et al., 1997), eluted as a single peak at 24.1 min on this
`column. The components of NC10 scFv-0 (VL–VH) protein
`were resolved by gel filtration on two Superose 12 HR10/30
`columns linked in tandem, yielding two protein peaks with
`apparent molecular masses of ~108 and ~78 kDa (Figure 3b).
`In contrast to NC10 scFv-0 (VH–VL), the tandem gel filtration
`indicated that NC10 scFv-0 (VL–VH) forms not only the
`expected trimer (~78 kDa) but also a tetramer (~108 kDa).
`Attempts to isolate homogeneous NC10 scFv-0 tetramer were
`only partially successful, as isolated tetramer rapidly inter-
`converted into a mixture of tetramer and trimer (data not
`shown). This observation indicated a relatively rapid equilib-
`rium between NC10 scFv-0 VL–VH tetramer and trimer with
`the tetramer being the predominant species as shown in
`Figure 3.
`Expression of NC10 VL–VH scFvs with variable linker length
`To investigate further the effect of linker length upon multimer-
`ization of NC10 scFvs in reverse (VL–VH) orientation, seven
`different NC10 scFv constructs with linkers from 15 to 0
`amino acid residues were assembled in pGC secretion vector
`(Coia et al., 1997) as described in Materials and methods.
`This vector was chosen for construction and expression of
`these scFvs with shortened linkers in preference to pPOW
`
`UPenn Ex. 2062
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`O.Dolezal et al.
`
`Fig. 5. Size-exclusion chromatography on Superdex 200 HR 10/30 column
`of affinity-purified NC10 scFv VL–VH proteins. The column was
`equilibrated in PBS, pH 7.4, and the flow-rate was 0.5 ml/min. (a) Shows
`the scFv-5 (dotted line), scFv-4 (dashed line) and scFv-3 (solid line), all
`eluting predominantly as dimers; (b) shows scFv-15 (solid line) containing
`monomer and dimer, and scFv-0 (dashed line) eluting as a mixture of
`mainly tetramer and trimer; (c) shows the scFv-1 (dashed line) and scFv-2
`(solid line), each containing a mixture of dimers, trimers, tetramers and
`higher molecular mass multimers.
`
`Size-exclusion chromatographic profiles of scFv-1 and
`scFv-2 yielded a series of peaks consistent with an equilibrium
`mixture of scFv dimers, trimers, tetramers and higher molecular
`mass multimers (Figure 5c). In the case of scFv-2 this
`equilibrium mixture was biased towards the smaller scFv
`entities whereas in case of scFv-1 the equilibrium mixture
`shifted towards higher molecular mass multimers. Analysis of
`entire scFv-1 and scFv-2 protein mixtures in 60% ethylene
`glycol and at lower concentration (⬍100 µg/ml) showed that
`the higher molecular mass multimers dissociated primarily
`into dimers and trimers (data not shown). This suggests that
`the dimers and trimers of scFv-2 and particularly scFv-1 are
`
`570
`
`relatively unstable and as a result prefer to assemble into
`higher molecular mass multimers as the linker length is reduced
`from two to one amino acid residue.
`As described previously (Figure 1), when compared with
`the original NC10 scFv VH–VL proteins, a second FLAG
`sequence was included in our constructs to improve the
`purification efficiency of various NC10 VL–VH proteins. To
`confirm that this additional FLAG sequence does not affect
`the state of oligomerization, several pGC-NC10 VL–VH clones
`containing a single FLAG sequence were also constructed and
`expressed in E.coli. The ‘single-FLAG’ NC10 VL–VH proteins
`with no, one, two and 15 residue linkers were purified on an
`anti-FLAG affinity column and analysed by gel filtration
`chromatography. The resulting gel filtration profiles indicated
`no apparent difference in oligomerization properties between
`‘single-FLAG’ and ‘double-FLAG’ NC10 VL–VH proteins
`(data not shown). The presence of minor bands at ~26 kDa in
`affinity-purified NC10 VL–VH samples (Figure 4) provided
`further evidence that the additional FLAG peptides do not
`affect the oligomeric status of these proteins. These bands
`occur owing to a gradual cleavage of FLAG peptides from the
`C-terminus of NC10 scFv proteins during extended storage at
`4°C (demonstrated by Western blot analysis with the anti-
`FLAG M2 antibody, data not shown). The presence of these
`bands, however, had no significant effect on the gel filtration
`profiles, and hence multimeric forms, of these NC10 scFvs
`(data not shown).
`Formation of complexes with 3–2G12 anti-idiotype Fab⬘
`fragments
`To analyse further the multimeric status of NC10 scFvs
`(VL–VH), the Fab⬘ fragment of 3–2G12 anti-idiotype mono-
`clonal antibody, which competes with influenza virus neura-
`minidase for binding to NC10, was used to form complexes
`with various scFv VL–VH proteins. Affinity-purified scFv-5
`dimer and scFv-5 tetramer, scFv-0 tetramer as well as the
`scFv-2 and scFv-1 mixtures were complexed with 3–2G12
`Fab⬘ fragment and the resulting complexes analysed by gel
`filtration (data not shown) and by electron microscopy (see
`below). In the case of scFv-5,
`the tetramer species was
`separated from dimer by gel filtration just prior to binding to
`3–2G12 Fab⬘. Complexes were formed by mixing dimers (for
`scFv-5) and tetramers (for scFv-5 and scFv-0) in a 1:2 and
`1:4 molar ratio, respectively, with the 3–2G12 Fab⬘ fragment.
`The Fab⬘ was kept in slight excess to these ratios to ensure
`complete decoration of all antigen binding sites present. The
`resulting scFv dimer–Fab⬘ and scFv tetramer–Fab⬘ complexes
`were then purified by size-exclusion chromatography on a
`Superdex 200 column (data not shown). There was no evidence
`of unbound scFv tetramer (for scFv-0 and scFv-5) or scFv
`dimer (for scFv-5) in any of these preparations. The scFv-5
`dimer–Fab⬘ complex eluted at an elution time corresponding
`to a molecular mass of ~160 kDa, which is consistent with
`the mass of a complex of two Fab⬘ molecules (~52 kDa each)
`and one scFv-5 dimer (~55 kDa). The scFv tetramer–Fab⬘
`complex (both scFv-0 and scFv-5) eluted at an elution time
`corresponding to a molecular mass of ~320 kDa, which is
`consistent with the mass of a complex of four Fab⬘ molecules
`and one scFv tetramer (~108 kDa). The estimated molecular
`mass of scFv–anti-idiotype Fab⬘ complexes is consistent with
`the prediction that scFv-5 dimers are bivalent and bind two
`Fab⬘ fragments and that scFv-0 (and scFv-5) tetramers are
`tetravalent and bind four Fab⬘ fragments. Minor peaks were
`
`UPenn Ex. 2062
`Miltenyi v. UPenn
`IPR2022-00855
`
`

`

`scFv multimers NC10scFVmultimers
`
`Fig. 6. Electron micrographs of scFv–3–2G12 Fab⬘ complexes stained with 2% potassium phosphotungstate. Magnification bar is 50 nm. (a) Field of scFv-5
`dimer complexes. (b) Field of scFv-5 tetramer complexes. (c) Field of scFv-0 tetramer complexes. At least one scFv-0 triabody (circled) is also present.
`(d) Field showing projections of compl

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