throbber
doi:10.1016/j.jmb.2003.10.040
`
`J. Mol. Biol. (2004) 335, 41–48
`
`COMMUNICATION
`Optimization of the Antibody CH3 Domain by Residue
`Frequency Analysis of IgG Sequences
`
`Stephen J. Demarest*, Jeff Rogers and Genevie` ve Hansen
`
`Torrey Mesa Research Institute
`3115 Merryfield Row, San
`Diego, CA 92130, USA
`
`In an attempt to enhance the overall assembly, yield and half-life of
`recombinant antibody proteins, we have cloned and expressed several
`IgG1 CH3 domains and examined their folding/refolding characteristics.
`We utilized a cytoplasmic bacterial expression system with a thioredoxin
`reductase knock-out strain of BL21(DE3) to produce bovine, murine and
`human CH3. Under identical conditions, expression of bovine CH3 resulted
`consistently in the highest yields of properly folded/oxidized protein.
`Circular dichroism and fluorescence experiments demonstrate that
`oxidized bovine and murine CH3 have surprisingly similar structures
`and stabilities, considering the marginal sequence conservation between
`the two molecules. Residue frequency analysis using a limited data set of
`36 unique Fc sequences originating from 19 different mammalian species
`targeted five specific sites for optimization within bovine CH3. Combi-
`nation of three of these mutants increased the thermal stability of the
`molecule to 86 8C. Comparison of this approach to similar studies using
`larger sequence databases and/or different selection criteria suggests
`sequence database design can increase the success rate for identifying
`residue sites worth optimizing. This optimized CH3 domain can be used
`as a particularly stable platform for functional design and can be grafted
`into full-length antibody sequences to enhance their thermodynamic
`parameters and shelf-life.
`
`q 2003 Elsevier Ltd. All rights reserved.
`
`*Corresponding author
`
`Keywords: antibody; immunoglobulin; CH3; protein design; protein folding
`
`antibody-based pharmaceuticals
`Monoclonal
`represent a large and growing fraction of all drug
`candidates currently entering clinical trials.1 Cur-
`rent technologies have enabled the production of
`antibodies capable of recognizing virtually any
`antigen or molecular target with extremely high
`
`Supplementary data associated with this article can be
`found at doi: 10.1016/y.jmbi.2003.10.040
`Present addresses: S. J. Demarest, J. Rogers, G. Hansen,
`Diversa Corp., 4955 Directors Place, San Diego, CA
`92121, USA.
`Abbreviations used: ASA, accessible surface area;
`bCH3, bovine CH3; Fc, antibody heavy chain CH2CH3
`dimer; Hepes, 4-(2-hydroxyethyl)-1-
`piperazineethanesulfonic acid; LB, Luria broth; hCH3,
`human CH3; mCH3, murine CH3; NCBI, National Center
`for Biotechnology Information; scFv, single-chain
`antibody variable domains; Tm, temperature midpoint of
`thermal denaturation.
`E-mail address of the corresponding author:
`stephen.demarest@diversa.com
`
`affinity. Additionally, antibodies can identify and
`neutralize pathogens (bacteria, viruses),
`toxins
`and cancerous cell types. Thus, antibodies have
`emerged as powerful diagnostic tools in all aspects
`of life sciences, as well as an attractive means of
`creating highly specific therapeutics.
`Mass production of antibodies is costly and
`requires tedious processing before the product is
`ready for market.2 For these reasons, many recom-
`binant methodologies for producing monoclonal
`antibodies are being investigated, including pro-
`duction in mammalian, plant, yeast and bacterial
`cell systems.3,4 The ability to increase the yield and
`molecular half-life of fully assembled and func-
`tional antibody products within these expression
`systems is also a major concern.5
`Here, we describe our approach to optimize the
`portion of
`the IgG Fc responsible for heavy
`chain association (i.e. the CH3 domain; Figure 1).
`Traditionally, research has focused on the study of
`antibody variable domains to understand their
`
`0022-2836/$ - see front matter q 2003 Elsevier Ltd. All rights reserved.
`
`Exhibit 2070
`Page 01 of 08
`
`

`

`42
`
`Antibody Constant Domain Folding and Optimization
`
`Figure 1. Top: A ribbon diagram of the X-ray crystal structure of human CH3.20,39 Residue positions identified as
`potential sites for optimization on the basis of residue frequency analysis are displayed in stick format. The ribbon dia-
`gram was rendered using MolMol.32 Middle: Alignment of the consensus sequence from a non-redundant database of
`36 NCBI mammalian IgG Fc sequences against the human, bovine and murine IgG1 CH3 sequences. The human CH3
`domain was cloned from Genbank source #BC026038 purchased from the ATCC. Bovine CH3 was cloned from
`in-house bovine constructs derived from a bovine spleen cDNA library purchased from Stratagene. Murine CH3 was
`cloned from an in-house synthetic murine IgG1 gene based on the murine IgG1 Kabat database consensus sequence.33
`CH3 PCR inserts and commercial pET21a plasmid (Novagen) were digested using the designed Bam HI and Xho I
`restriction sites (enzymes from Invitrogen). Inserts were ligated into the vector using the H.C. T4 DNA ligase (Invitro-
`gen). The QuickChangew Site-Directed Mutagenesis kit (Stratagene) was used to create mutations in the bovine CH3
`pET21a vector. Residue positions within the bovine CH3 domain where directed mutations were made are indicated.
`Below the alignments, (.), conservative variation; (:), strict variation; (*), no variation. All inserts and mutations were
`confirmed by DNA sequencing. BL21-trxB(DE3) (Novagen) was utilized as the host for IPTG-inducible cytoplasmic
`expression. BL21(DE3) did not yield significant amounts of oxidized protein and Rosetta-Gami (Novagen) had a
`significantly longer growth phase without providing a significant increase in CH3 oxidation over the BL21-trxB(DE3)
`
`Exhibit 2070
`Page 02 of 08
`
`

`

`Antibody Constant Domain Folding and Optimization
`
`43
`
`antigen-binding capabilities.6 However, effector
`functions
`that
`allow antibodies
`to
`involve
`additional factors within the immune system are
`embedded within the constant domains.7,8 For
`these reasons, the past few years have witnessed
`an explosion in the sequencing of antibody con-
`stant domains. We utilize the publicly available Fc
`sequences to find residue positions within the
`bovine IgG1 CH3 fold that are poorly represented
`and describe a limited set of mutations at these
`sites designed to enhance the thermal stability of
`the domain.
`The CH3 domain of bovine IgG1 was chosen as
`our model system on the basis of analysis of a con-
`densed set of 36 IgG Fc
`sequences. These
`sequences were collected by performing the Tera-
`Blaste routine on the non-redundant protein
`sequence database maintained by NCBI
`(The
`National Center for Biotechnology Information)
`using bovine, murine and human Fc sequences as
`bait. Other than primate IgG sequences, bovine
`IgG1 demonstrated the closest resemblance to the
`consensus sequence, while exhibiting a few defini-
`tive sites that, once mutated, might
`lead to
`increases in thermal stability. The sequences of
`bovine, human and murine IgG1 CH3 (denoted
`bCH3, hCH3 and mCH3, respectively) are shown in
`Figure 1. Proteins with disulfides have tradition-
`ally been expressed in the periplasmic space of
`bacteria.38 However, cytosolic yields tend to be
`much higher; therefore, bCH3 was expressed in the
`cytoplasm of several bacterial cell lines. The best
`results were obtained in BL21-trxB(DE3),
`a
`bacterial cell line carrying a mutation in the thiore-
`doxin reductase gene, trxB.10 bCH3 was expressed
`at 20 – 40 mg/l in a shaker flask in BL21-trxB(DE3).
`The protein was affinity-purified on Ni2þ
`-NTA
`resin (Qiagen) using the C-terminal hexahistidine
`tag. Oxidized protein was separated from reduced
`material using reverse-phase HPLC (Figure 1).
`Purified yields of folded/oxidized protein were
`approximately 70% of the expressed material. The
`exact mass of oxidized bCH3, 15064.9402 AMU,
`was determined by electrospray,
`time-of-flight
`(TOF) mass spectroscopy. The reduced fraction
`(analyzed in the presence of DTT) yielded a major
`peak with a molecular mass of 15066.9403 AMU,
`exactly 2 AMU higher than the oxidized form, due
`to the protonation of the free thiol groups. The
`theoretical masses of oxidized and reduced bCH3
`are 15066.6 and 15068.6, respectively, and are indis-
`tinguishable within the error of the measurement
`from the experimentally derived values. Oxidized
`mCH3 could be obtained in only small quantities
`
`(, 1 mg per 1 l shaker flask) and oxidized hCH3
`could not be separated from its reduced form
`using our purification scheme.
`The CH3 domain is known to form a specific non-
`covalent dimer.11 Oxidized bCH3 runs appro-
`priately as a dimer on a BioSpec2000 (Pharmacia)
`HPLC gel-filtration column. Molecular mass stan-
`dards included bovine serum albumin (66 kDa),
`carbonic anhydrase (28 kDa),
`lysozyme (14 kDa)
`and ubiquitin (8.5 kDa). The elution time of bCH3
`was constant over a 200 nM – 200 mM injected
`range of concentrations. Similar to bCH3, oxidized
`mCH3 appears to be a dimer, as judged by gel-
`filtration. Purified hCH3 elutes in several forms,
`predominantly trimer, dimer and monomer, an
`indication that it is not wholly folded/oxidized.
`Repeated expressions/purifications of hCH3 never
`produced viable protein.
`three CH3 domains exhibited significant
`All
`secondary structure, as judged by their circular
`dichroism (CD) spectra (Figure 2A). Far-UV CD
`spectra of bCH3 solubilized from cell pellets using
`8 M urea and subsequently Ni2þ
`-purified in urea
`and refolded were identical with the bCH3 spectra
`solubilized and purified using strictly aqueous
`buffers. bCH3, mCH3 and hCH3 all have minima at
`217 nm, a characteristic of b-sheet structures con-
`sistent with the antibody fold being composed of
`a twofold b-sheet sandwich. The hCH3 spectrum
`was significantly different in intensity and shape
`from the spectra of bCH3 and mCH3, but was very
`similar to what was observed for reduced bCH3
`the bCH3 and
`(data not shown). Surprisingly,
`mCH3 spectra were nearly superimposable, indicat-
`ing that their structures must be nearly identical.
`This was unexpected considering bCH3 and mCH3
`are only 54% identical in sequence.
`the CH3
`Temperature-induced unfolding of
`domains was monitored separately using fluor-
`escence, near-UV CD and far-UV CD. The fluor-
`escence excitation wavelength was 278 nm with a
`3 nm bandwidth and no emission cutoff filter; the
`near-UV CD signal was monitored at 278 nm
`using a 2 nm bandwidth; and the far-UV CD signal
`was monitored at 217 nm using a 1 nm bandwidth.
`bCH3 and mCH3 undergo single unfolding tran-
`sitions with midpoints (Tm) at 348.8(^ 0.5) K and
`346.8(^ 0.5) K,
`respectively. Refolding of both
`bCH3 and mCH3 was reversible. The CD spectra at
`278 K (5 8C) prior to and one hour after tempera-
`ture-denaturation were virtually unchanged.
`The Tm of both bCH3 and mCH3 were unaffected
`by concentration. Fluorescence measurements
`were performed using 0.5 mM and 4 mM protein,
`
`strain. Bottom: Purification of bCH3. Bacterial pellets containing CH3 constructs were dissolved in 8 M urea (pH 8),
`spun down and the supernatants were passed over Ni2þ
`-NTA resin (Qiagen). Protein was eluted from the resin by
`adding 8 M urea (pH 4). Reverse-phase HPLC was performed on a Dionex DX500 chromatography system using a
`Phenomenex C5-Prep column with water/acetonitrile gradients and an absorbance detector set to 280 nm. The bottom
`panel displays the chromatograph of bCH3. The peak eluting at 22.9 minutes is oxidized CH3, while the peak eluting at
`25.4 minutes is reduced CH3.
`
`Exhibit 2070
`Page 03 of 08
`
`

`

`44
`
`Antibody Constant Domain Folding and Optimization
`
`range of concentrations, no change in the Tm was
`observed, as might be expected for a folded dimer
`transition.12
`to unfolded monomer
`two-state
`Buchner and co-workers, however, observed a con-
`centration-dependent change in the guanidinium –
`HCl denaturation midpoint of the CH3 domain
`from the murine monoclonal antibody MAK33
`typical for a native dimer to unfolded monomer
`transition.13,14 Thermal unfolding transitions of the
`dimeric Arc Repressor protein and the dimeric
`four-helix bundle protein, ROP, both display con-
`centration-dependent Tm shifts.15,16 The lack of a
`temperature shift between different concentrations
`of protein could be explained if the folding rate of
`the protein is not dominated by the rate of dimer
`association, but instead dominated by the trans –
`cis isomerization of P177(374). Another possibility
`could be that the dimer dissociates before reaching
`the temperature at which the protein unfolds. Due
`to the uncertainty in the model that should be
`applied for quantitative interpretation of the free
`energy of folding, we use the Tm value of bCH3 as
`a standard to assess the relative stability changes
`induced by the designed mutants described below.
`Our goal was to discover residue positions
`fully optimized for protein
`within bCH3 not
`stability. We performed positional frequency and
`positional entropy analyses on our limited IgG
`data set (36 sequences from 19 mammalian species)
`utilizing a Perl program written in-house. A
`ClustalW alignment of
`the 36 sequences
`is
`provided in the Supplementary Material†.17 All
`sequences were at least 95% unique (i.e. at least
`ten residue variations out of , 220 residues from
`any other sequence within the data set). Without
`the 5% cutoff, the dataset burgeons to over 100
`sequences. The redundancy creates considerable
`bias and is therefore eliminated. The residue fre-
`quency, piðrÞ; for each position, i, in an individual
`sequence is simply the number of times that a
`(r ¼ A,C,D…V,W,Y)
`particular
`residue-type
`is
`observed within the dataset divided by the total
`number of sequences. The positional entropy, NðiÞ;
`was calculated as a measure of every residue
`position’s variability.18,19 The positional entropy is
`a function of the Shannon informational entropy,
`HðiÞ :
`
`XY
`
`r¼A
`
`NðiÞ ¼ eHðiÞ
`
`; HðiÞ ¼ 2
`
`piðrÞlnðpiðrÞÞ
`
`ð1Þ
`
`Data from the residue frequency analysis were
`used to design mutants capable of optimizing
`bCH3 stability. Mutations were selected based on
`four criteria. The first is that the bovine sequence’s
`residue frequency at a given position is much
`smaller than the most common residue frequency,
`mcpiðrÞ; (i.e. piðrÞ=mcpiðrÞ # 0:2). Next, mutations
`must be conservative based on CH3 structure
`analysis and residue type. Thus, no charge reversals
`
`† http://www.ebi.ac.uk/clustalw
`
`Figure 2. Structure and stability analyses of bCH3,
`mCH3 and hCH3. A, The CD spectra of bCH3, mCH3 and
`hCH3 at 5 8C. The CD and fluorescence measurements
`were performed using an Aviv model 215 spectrometer
`equipped with a thermoelectric cuvette-holder and the
`total fluorescence accessory. The buffer contained 2 mM
`phosphate, borate, citrate, 10 mM NaCl (pH 7.5). All
`final spectra were the average of at least four scans
`utilizing a signal averaging time of 2 s/l. Signal
`averaging times for all melting experiments were 50 s/
`deg. C. Protein concentrations were determined using
`the method of Pace and co-workers.34 B. Temperature-
`denaturation of bCH3, mCH3 and hCH3 monitored by
`the far-UV CD signal at 217 nm. All melts increased the
`temperature at 2 deg. C intervals for CD and fluor-
`escence sampling and utilized a 180 s equilibration
`period between data acquisitions. The melting tempera-
`ture of bovine and murine CH3 did not vary with concen-
`tration; therefore the far and near-UV CD curves and the
`fluorescence curves were all fit to a two-state, single-
`molecule unfolding model.35 The stability of each variant
`was qualitatively ranked using Tm values. A theoretical
`DCp8, 1727 cal/mol K (1 cal ¼ 4.184 J), was calculated
`based on an estimate of the change in accessible surface
`area (DASA) between the folded and unfolded states of
`the CH3 dimer.36,37
`
`far-UV CD measurements were performed using
`10 mM protein and near-UV CD measurements
`using 30 mM protein (see Figure 2B for far-UV
`CD temperature-denaturation). Over this 60-fold
`
`Exhibit 2070
`Page 04 of 08
`
`

`

`Antibody Constant Domain Folding and Optimization
`
`45
`
`bovine IgG2 or IgG3). This final criterion is related
`to the no-covariation criterion.
`Five potentially non-ideal residue sites were
`identified when pitting the bovine CH3 sequence
`against the Fc data set. These positions have been
`rendered onto the structure in Figure 1 and are
`indicated in the sequence alignment below the
`structure. Several CH3 domains, including bovine
`CH3, contain insertions or deletions resulting in an
`ambiguous numbering system when compared to
`the human IgG sequence. Therefore, we chose to
`number according to our Fc data set with residue
`G35 corresponding to residue G237, the first resi-
`due of the Fc structure of human IgG1.20 Numbers
`in parentheses refer to the standard full-length
`human IgG residue numbering, while the numbers
`directly to the left of the residue letter refer to num-
`bering originating from our Fc data set. The sites
`are S174(371), Y179(376), G197(392), S207(402) and
`T246(441). Each residue site is spatially distant
`from the four other sites, on the basis of the struc-
`ture of hCH3,20 and is thus expected to have an
`independent and additive effect on the stability of
`the domain. The following point mutations were
`made based on the residue frequency analysis of
`bCH3: S174G, Y179D, G197K, G197A (conservative
`mutation), S207G and T246L. Four of these five
`mutant positions are at residue sites of reasonable
`heterogeneity
`(i.e.
`the measured
`positional
`entropies are in the top 70% for all residue pos-
`itions). Mutation at S207(402) was the only position
`that was highly conserved; only four of the 36
`sequences have a residue other than Gly, with Ser
`being represented twice.
`All mutant proteins display nearly identical CD
`spectra compared to the native protein (Figure 3A),
`indicating that no major structural change was
`induced by any of
`the point mutations. Each
`mutant domain exhibits a reproducible Tm, as
`judged by both near and far-UV CD (Figure 3B).
`Near and far-UV CD denaturations were per-
`formed on all the mutants, using the same concen-
`trations that were used to study the wild-type
`protein (Table 1). G197K, S207G and T246L
`mutations had a stabilizing affect on bCH3. Combi-
`nation of these mutants into a single construct led
`to near-additive increases in the thermal stability
`of
`the domain. The experimentally determined
`
`Figure 3. Structure and stability analyses of bCH3 and
`the mutant constructs of the domain. A, The CD spectra
`of native and mutant bCH3 constructs at 5 8C, pH 7.5.
`B, Plots of the unfolded fractions of the bCH3 variants
`between 325 and 375 K.
`
`were made and no large, buried hydrophobic
`amino acids were converted to charged residues
`or Gly. The third criterion is that there must be an
`absence of co-variation at this position with other
`residue positions throughout the sequence. Lastly,
`the residue to be mutated must not be conserved
`within the individual species IgG subclass (i.e. in
`
`Table 1. Results of fitting the CD temperature melts of bCH3, the six single-mutant variants and a triple-mutant variant
`to a two-state unfolding model
`
`Construct
`
`Tm (K) fluor. Ex. l280
`
`Tm (K) CD [u]217
`
`Tm (K) [u]280
`
`Tm (K) Avg.
`
`DTm (K)
`
`bCH3
`S174G
`Y179D
`G197K
`G197A
`S207G
`T246L
`G197K/S207G/T246L
`pG197K/S207G/T246L
`
`348.0 ^ 0.3
`–
`–
`–
`–
`–
`–
`–
`–
`
`349.9 ^ 0.3
`348.7
`349.3
`354.2
`351.2
`352.6
`352.2
`–
`–
`
`349.0 ^ 0.3
`349.6
`349.9
`354.2
`352.2
`353.7
`352.3
`358.8
`–
`
`348.8 ^ 0.5
`349.1
`349.6
`354.2
`351.7
`353.2
`352.3
`358.8
`p361.1
`
`–
`0.3
`0.8
`5.4
`2.9
`4.4
`3.5
`10.0
`p13.3
`
`The asterisk ( p ) indicates a hypothetical triple mutant, assuming the individual mutant stabilizations are additive.
`
`Exhibit 2070
`Page 05 of 08
`
`

`

`46
`
`Antibody Constant Domain Folding and Optimization
`
`increase in Tm was slightly lower than the increase
`predicted when assuming full additivity; however,
`the difference between the two numbers lies at the
`edge of experimental uncertainty. Temperature-
`denaturation curves of bCH3, the six mutant bCH3
`domains, and the experimental and hypothetical
`triple mutant are shown in Figure 3B.
`The stabilizing affect of the three mutations can
`be rationalized by analyzing the structure of the
`hCH3 domain.20 G197(392)K inserts a Lys at the
`interface between the CH3 dimer. The hydrophobic
`portion of the side-chain packs against a hydro-
`phobic patch created by F210(405) and V202(397)
`of the opposite dimer subunit. Adding a hydro-
`phobic moiety to this cavity also explains why Ala
`is preferred over Gly at this position. The posi-
`tively charged primary amine is exposed to solvent
`and is in close proximity to D204(399), possibly
`enhancing the favorability of Lys at this position
`over Ala. S207(402)G places glycine in a tight turn
`between b-strands. The backbone dihedral angles
`of this residue in the crystal structure are outside
`the generally allowed ranges for a-substituted
`amino acids (all amino acids other than Gly).
`Mutation to Gly at this position likely releases
`strain induced by the tight
`turn. Finally,
`the
`T246(441)L mutation completely buries the iso-
`butyl side-chain into the hydrophobic interior of
`the b-sandwich. The favorable contribution is not
`as large as might be expected for the burial of an
`additional three methylene groups, indicating that
`steric placement or hydrogen bonding of the Thr
`side-chain must compensate somewhat
`for the
`loss of buried hydrophobic surface area.21
`The expression levels of the mutant variants
`were relatively unchanged compared to that of the
`wild-type bCH3. Variants with enhanced thermal
`stability did not have
`increased expression
`compared to the wild-type protein. One way of
`rationalizing this result is that the thermal stability
`of the native protein, 76 8C, is so far above 37 8C,
`the temperature at which the bacteria were
`cultured,
`that
`stabilization does not
`affect
`expression significantly. There was one exception,
`S174G. Expression of this variant led to protein
`yields fourfold higher than what was observed for
`wild-type bCH3 and the other variants. The
`mutation had a minimal effect on the overall stab-
`ility of the domain. It is possible that the S174G
`mutation leads to increased kinetics of folding and
`therefore less in vivo degradation of the protein.
`Overall, however, increased thermal stability did
`not correlate with increased expression.
`Many approaches have been applied success-
`fully to increase the thermal stability of proteins
`or enzymes, including rational design,22 random
`mutagenesis,23 computational design,24 disulfide
`engineering25 and even the creation of circular
`proteins.26 Statistical analysis of protein sequence
`data sets provides another rapid and effective
`means of semi-rationally designing more stable
`proteins, provided a big enough data set exists
`from which to draw information. Davidson and
`
`co-workers have used residue frequency analysis
`using a database of 266 non-redundant SH3
`domain sequences to stabilize the C-terminal SH3
`domain of
`the yeast actin-regulating protein
`Abp1p.18,27 This , 60 residue protein is only
`marginally stable and contains many non-standard
`SH3-type residues. Their mutational criteria were
`similar to those reported here; however, four of
`their seven mutations had a destabilizing effect on
`the domain, whereas none of the bCH3 mutations
`was destabilizing. Bovine IgG1 matches the IgG
`consensus sequence to a much greater extent than
`Abp1p matches the SH3 consensus; approximately
`50% of the Abp1p sequence did not match the con-
`sensus. The magnitude of the stabilizing mutations
`within Abp1p led were much greater, however,
`increasing the Tm values on average by 12 deg.C.
`This may be due to the greater size of their data-
`base and its ability to statistically find the real
`winner at an individual position. Also, the low
`intrinsic stability of
`the Abp1p domain likely
`leaves room for greater enhancement. The pitfall
`of choosing residues for replacement based on the
`statistically most frequent residue type is that each
`particular
`residue position interacts with and
`directs the best residue types of residues around
`it.28 However, the limited taxonomy of the mam-
`malian Fc database described here likely does not
`allow for
`the broad evolutionary sampling
`required to generate significant linked pairs of
`mutations.
`Antibody sequencing efforts that have focused
`primarily on antibody variable domains have pre-
`viously enabled residue frequency analysis to aid
`in the design of a thermostable variable domain.29
`Steipe and co-workers utilized all the heavy-chain
`variable domain sequences of the Kabat database
`to design mutations at several positions: six of ten
`mutants were stabilizing, three had no effect on
`stability and only one was destabilizing. Their
`relatively high success rate, similar to ours, may
`be due, in part, to the limited taxonomy of their
`database allowing them to avoid significant
`divergence between sequences,
`leading to more
`instances of covariation. The number of mam-
`malian IgG constant domains that have been
`sequenced is comparatively small. Only recently,
`with the number of publicly available unique IgG
`constant domains tripling in the last five years,
`has residue frequency analysis of the IgG constant
`domains been possible.
`Although the strict criteria used here strength-
`ened our chances of choosing stabilizing mutations
`successfully, they almost certainly exclude other
`potentially stabilizing mutants that do not fall
`within the limits.
`In two independent studies,
`(using GroEL)30
`and
`Wang and co-workers
`Nikolova and co-workers (using the p53 DNA-
`binding domain)9 utilized a different
`residue
`frequency strategy to find residue positions worth
`optimization. Their criterion for mutation was to
`create libraries at residue sites with high positional
`entropies rather than adopting our strategy of
`
`Exhibit 2070
`Page 06 of 08
`
`

`

`Antibody Constant Domain Folding and Optimization
`
`47
`
`mutating residue positions where the wild-type
`residue frequency was extremely low.
`In both
`reports, residue sites of improvement were found
`where the wild-type residue frequency was as
`high as 20 – 91% of all the residues observed for
`those individual positions. Our strategy ignores
`potentially stabilizing mutations such as these.
`The positive aspect of mutating at positions of low
`residue frequency was that five of six residue
`replacements stabilized the domain and none was
`destabilizing. The success rate for finding posi-
`tively stabilizing mutants was much lower in the
`designed mutants of GroEL and p53.
`In both
`studies, approximately 29% of the mutations were
`stabilizing, 42% had relatively little affect on the
`stability and 29% were destabilizing.
`Biophysical studies of the CH3 domain have
`focused mainly on the human and murine
`proteins.13,31,32 The Tm values of bovine CH3 and
`murine CH3 measured here were both close to the
`reported Tm of the human domain, 80.4 8C.32 Both
`the bovine and murine domains refolded to their
`native structures within an hour of lowering the
`temperature to favor the native state. This is in
`fair agreement with the work of Buchner and
`co-workers. At 4 8C, they observe approximately
`80% refolding of their murine CH3 construct over
`the period of an hour, with the accumulation of a
`folding intermediate accounting for the other 20%
`of the population. However, we refolded while
`cooling from 101 8C over a period of an hour. The
`native state becomes stable at temperatures below
`75 8C, where folding/refolding is likely to be
`accelerated greatly and it is not surprising that we
`observe nearly full recovery of folded protein over
`the course of an hour.
`Creating a stabilized CH3 or Fc domain as
`described here would be useful as a platform for
`functional design. Potential reasons for function-
`ally modifying constant domains include modify-
`ing the interaction strengths of Fc domains for
`individual Fc receptors8 or for the creation of
`bispecific
`antibodies. Carter
`and co-workers
`designed a heterodimeric CH3 system with the
`goal of producing antibodies capable of binding
`two targets.32 They achieved remarkable success;
`however,
`their
`initial heterodimeric constructs
`were
`significantly
`compromised in stability
`compared to the wild-type protein. Functional
`mutations often lead to decreases in the stability
`of a protein. Combining these mutations can
`destabilize the fold to a point where most of the
`protein remains unfolded and functionless. Start-
`ing from an ultra-stable platform would provide
`more room for compromise when mutating for
`functional purposes.
`In conclusion, we demonstrated that the publicly
`available IgG sequences, although limited in
`number,
`provide
`enough
`information
`for
`sequence-based optimization of CH3 stability. The
`limited taxonomy of the database appeared to
`enhance
`our
`ability
`to pinpoint
`stabilizing
`mutations, suggesting that database design may
`
`increase the success rate for identifying residue
`positions that can be optimized. Future studies
`may include the engineering of thermostable, full-
`length antibody constructs with increased half-life
`and potentially increased molecular yield.
`
`Acknowledgements
`
`The authors thank G. Chen for critically reading
`the manuscript and for insightful discussions.
`
`References
`
`1. van Dijk, M. A. & van de Winkel, J. G. J. (2001).
`Human antibodies as next generation therapeutics.
`Curr. Opin. Chem. Biol. 5, 368 – 374.
`2. Gura, T. (2002). Therapeutic antibodies: magic bullets
`hit the target. Nature, 417, 584 – 586.
`3. Verma, R., Boleti, E. & George, A. J. T. (1998). Anti-
`body engineering: comparison of bacterial, yeast,
`insect
`and mammalian
`expression
`systems.
`J. Immunol. Methods, 216, 165 – 181.
`4. Smith, M. D. (1996). Antibody production in plants.
`Biotechnol. Advan. 14, 267 –281.
`5. Wo¨ rn, A. & Plu¨ ckthun, A. (2001). Stability engineer-
`ing of antibody single-chain Fv fragments. J. Mol.
`Biol. 305, 989 –1010.
`6. Muyldermans, S. (2001). Single domain camel anti-
`bodies: the current status. J. Biotechnol. 74, 277 –302.
`7. Presta, L. G.
`(2002). Engineering antibodies for
`therapy. Curr. Pharm. Biotechnol. 3, 237 – 256.
`8. Ravetch, J. V. & Bolland, S. (2001). IgG Fc receptors.
`Annu. Rev. Immunol. 19, 275 – 290.
`9. Nikolova, P. V., Henckel, J., Lane, D. P. & Fersht, A. R.
`(1998). Semirational design of active tumor suppres-
`sor p53 DNA binding domain with enhanced
`stability. Proc. Natl Acad. Sci. USA, 95, 14675 – 14680.
`10. Proba, K., Ge, L. & Plu¨ ckthun, A. (1995). Functional
`antibody single-chain fragments from the cytoplasm
`of Escherichia coli: influence of thioredoxin reductase
`(TrxB). Gene, 159, 203 – 207.
`11. Isenman, D. E., Lancet, D. & Pecht, I. (1979). Folding
`pathways of immunoglobulin domains. The folding
`the Cg3 domain of human IgG1.
`kinetics of
`Biochemistry, 18, 3327 – 3336.
`12. Neet, K. E. & Tim, D. E. (1994). Conformational
`stability of dimeric proteins: quantitative studies by
`equilibrium denaturation. Protein Sci. 3, 2167 – 2174.
`13. Thies, M. J. W., Mayer, J., Augustine, J. G., Frederick,
`C. A., Lilie, H. & Buchner, J. (1999). Folding and
`association of
`the antibody domain CH3: prolyl
`isomerization preceeds dimerization. J. Mol. Biol.
`293, 67 –79.
`14. Lilie, H., Lang, K., Rudolph, R. & Buchner, J. (1995).
`Association of antibody chains at different stages of
`folding: prolyl
`isomerization occurs
`after
`the
`formation of quarternary structure. J. Mol. Biol. 248,
`190 –201.
`(1989). Equilibrium
`15. Bowie,
`J. U. & Sauer, R. T.
`the Arc repressor
`dissociation and unfolding of
`dimmer. Biochemistry, 28, 7139 – 7143.
`16. Steif, C., Weber, P., Hinz, H-J., Flossdorf, J., Cesareni,
`G. et al.
`(1993). Subunit
`interactions provide a
`significant contribution to the stability of the dimeric
`
`Exhibit 2070
`Page 07 of 08
`
`

`

`48
`
`Antibody Constant Domain Folding and Optimization
`
`four-a-helical-bundle protein ROP. Biochemistry, 32,
`3867 – 3876.
`17. Higgins, D., Thompson, J., Gibson, T., Thompson,
`J. D., Higgins, D. G. & Gibson, T. J. (1994). CLUSTAL
`W: improving the sensitivity of progressive multiple
`sequence alignment
`through sequence weighting,
`position-specific gap penalties and weight matrix
`choice. Nucl. Acids Res. 22, 4673 – 4680.
`18. Larson, S. M. & Davidson, A. R. (2000). The identifi-
`cation of conserved interactions within the SH3
`domain by alignment of sequences and structures.
`Protein Sci. 9, 2170 – 2180.
`19. Shenkin, P. S., Erman, B. & Mastrandrea, L. D. (1991).
`Information-theoretical entropy as a measure of
`sequence variability. Proteins: Struct. Funct. Genet. 11,
`297 – 313.
`20. DeLano, W. L., Ultsch, M. H., de Vos, A. M. & Wells,
`J. A. (2000). Convergent solutions to binding at a
`protein – protein interface. Science, 287, 1279 – 1283.
`21. Privalov, P. L. & Gill, S. A. (1988). Stability of protein
`structure
`and hydrophobic
`interaction. Advan.
`Protein Chem. 39, 191 – 234.
`(1997). Stabilization of
`22. Lee, B. & Vasmatzis, G.
`protein structures. Curr. Opin. Biotechnol. 8, 423 – 428.
`23. Gershenson, A. & Arnold, F. H. (2000). Enzyme
`stabilization by directed evolution. Genet. E

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