30.12.2013 Views

Discovery of human antibodies against the C5aR target using ... - DCU

Discovery of human antibodies against the C5aR target using ... - DCU

Discovery of human antibodies against the C5aR target using ... - DCU

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

JOURNAL OF MOLECULAR RECOGNITION<br />

J. Mol. Recognit. 2005; 18: 327–333<br />

Published online 10 February 2005 in Wiley InterScience (www.interscience.wiley.com). DOI:10.1002/jmr.735<br />

<strong>Discovery</strong> <strong>of</strong> <strong>human</strong> <strong>antibodies</strong> <strong>against</strong> <strong>the</strong><br />

<strong>C5aR</strong> <strong>target</strong> <strong>using</strong> phage display technology<br />

Lili Huang*, Aaron K. Sato, Meena Sachdeva, Tony Fleming, Susan Townsend and Daniel T. Dransfield<br />

<strong>Discovery</strong> Research, Dyax Corp., 300 Technology Square, Cambridge, MA 02139, USA<br />

Phage display technologies have been increasingly utilized for <strong>the</strong> generation <strong>of</strong> <strong>the</strong>rapeutic, imaging and<br />

purification reagents for a number <strong>of</strong> biological <strong>target</strong>s. Using a variety <strong>of</strong> different approaches, we have<br />

developed <strong>antibodies</strong> with high specificity and affinity for various <strong>target</strong>s ranging from small peptides to large<br />

proteins, soluble or membrane-associated as well as to activated forms <strong>of</strong> enzymes. We have applied this<br />

approach to G-protein coupled receptors (GPCRs), <strong>of</strong>ten considered difficult <strong>target</strong>s for antibody <strong>the</strong>rapeutics<br />

and <strong>target</strong>ing. Here we demonstrate <strong>the</strong> use <strong>of</strong> this technology for <strong>the</strong> identification <strong>of</strong> <strong>human</strong> <strong>antibodies</strong><br />

<strong>target</strong>ing <strong>C5aR</strong>, <strong>the</strong> chemoattractant GPCR receptor for anaphylatoxin C5a. The N-terminal region<br />

(residues 1–31) <strong>of</strong> <strong>C5aR</strong>, one <strong>of</strong> <strong>the</strong> ligand binding sites, was syn<strong>the</strong>sized, biotinylated and used as <strong>the</strong> <strong>target</strong><br />

for selection. Three rounds <strong>of</strong> selection with our proprietary <strong>human</strong> Fab phage display library were<br />

performed. Screening <strong>of</strong> 768 isolates by phage ELISA identified 374 positive clones. Based on sequence<br />

alignment analysis, <strong>the</strong> positive clones were divided into 22 groups. Representative Fab clones from each<br />

group were reformatted into IgGs and tested for binding to <strong>C5aR</strong>-expressing cells, <strong>the</strong> differentiated U-937<br />

cells. Flow cytometric analysis demonstrated that nine out <strong>of</strong> 16 reformatted IgGs bound to cells. Competition<br />

with a <strong>C5aR</strong> monoclonal antibody S5/1 which recognizes <strong>the</strong> same N-terminal region showed that S5/1<br />

blocked <strong>the</strong> binding <strong>of</strong> positive cell binders to <strong>the</strong> peptide used for selections, indicating that <strong>the</strong> identified cell<br />

binding IgGs were specific to <strong>C5aR</strong>. These antibody binders represent viable candidates as <strong>the</strong>rapeutic or<br />

imaging agents, illustrating that phage display technology provides a rapid means for developing <strong>antibodies</strong><br />

to a difficult class <strong>of</strong> <strong>target</strong>s such as GPCRs. Copyright # 2005 John Wiley & Sons, Ltd.<br />

Keywords: <strong>antibodies</strong>; phage display; GPCRs; <strong>C5aR</strong><br />

Received 2 November 2004; revised 10 December 2004; accepted 15 December 2004<br />

INTRODUCTION<br />

Over <strong>the</strong> past decade, <strong>antibodies</strong>, mainly used as research<br />

reagents, have been developed into effective <strong>the</strong>rapeutic<br />

agents with broad applications in cancer, inflammation<br />

and infectious diseases. Up to now, <strong>the</strong>re are 15 monoclonal<br />

<strong>antibodies</strong> approved for clinical use, more than 70 candidates<br />

in late stage clinical trials, and greater than 1000 in<br />

preclinical development (for review, see Stockwin and<br />

Holmes, 2003). The majority <strong>of</strong> <strong>the</strong>se antibody <strong>the</strong>rapeutic<br />

candidates are derived from classical hybridoma technology.<br />

To reduce immunogenicity in <strong>human</strong>s, <strong>the</strong>se mouse<br />

monoclonal <strong>antibodies</strong> are <strong>of</strong>ten <strong>human</strong>ized. The development<br />

<strong>of</strong> <strong>human</strong> antibody-based phage display libraries and<br />

<strong>human</strong> immunoglobulin transgenic mice has allowed <strong>the</strong><br />

discovery <strong>of</strong> <strong>human</strong> <strong>antibodies</strong>. One <strong>human</strong> monoclonal<br />

antibody, adalimumab, a TNF inhibitor, has recently<br />

been approved for <strong>the</strong> treatment <strong>of</strong> rheumatoid arthritis.<br />

*Correspondence to: L. Huang, <strong>Discovery</strong> Research, Dyax Corp., 300 Technology<br />

Square, Cambridge, MA 02139, USA. E-mail: lhuang@dyax.com<br />

Abbreviations used: BSA, bovine serum albumin; <strong>C5aR</strong>, C5a receptor; ELISA,<br />

enzyme-linked immunosorbent assay; FITC, fluorescein isothiocyanate; FOI,<br />

fraction <strong>of</strong> input; GPCRs, G-protein coupled receptors; HRP, horseradish<br />

peroxidase; IRES, eukaryotic internal ribosome entry site; mAb, monoclonal<br />

antibody; ONCL, oligonucleotide-assisted cleavage and ligation; PBS,<br />

phosphate-buffered saline; PBST, phosphate-buffered saline, 0.1% Tween-20;<br />

PCR, polymerase chain reaction; RBS, ribosome binding site; RT, room<br />

temperature; TMB, tetramethylbenzidine.<br />

More <strong>human</strong> <strong>antibodies</strong> are currently under evaluation in<br />

clinical trials and pre-clinical development. The use <strong>of</strong> <strong>the</strong>se<br />

new technologies has greatly shortened <strong>the</strong> time to develop<br />

<strong>human</strong> <strong>antibodies</strong> with high specificity and affinity.<br />

Phage display has become increasingly used for <strong>the</strong><br />

identification <strong>of</strong> <strong>the</strong>rapeutic, affinity purification and imaging<br />

agents (for review, see Azzazy and Highsmith, 2002).<br />

In phage display libraries, peptides and proteins including<br />

<strong>antibodies</strong> are displayed on <strong>the</strong> surface <strong>of</strong> filamentous phage<br />

through phage coat proteins such as pIII. Target-specific<br />

binders can be selected from phage display libraries through<br />

bio-panning processes. We have developed a series <strong>of</strong> Fab<br />

displaying phage libraries along with a procedure for rapidly<br />

reformatting <strong>the</strong>se Fabs into fully <strong>human</strong> IgGs (Hoet et al.,<br />

2004; Jostock et al., 2004). Using this approach, we have<br />

developed fully <strong>human</strong> <strong>antibodies</strong> with high specificity and<br />

affinity for various <strong>target</strong>s ranging from small peptides to<br />

large proteins, both soluble and membrane-associated, as<br />

well as to activated forms <strong>of</strong> enzymes. Here we demonstrate<br />

<strong>the</strong> use <strong>of</strong> this technology for <strong>the</strong> identification <strong>of</strong> <strong>human</strong><br />

<strong>antibodies</strong> <strong>target</strong>ing <strong>the</strong> C5a receptor (<strong>C5aR</strong>), a member <strong>of</strong><br />

<strong>the</strong> GPCR superfamily.<br />

GPCRs are seven-transmembrane G protein-coupled receptors<br />

involved in signal transduction cascades, contributing<br />

to many biological activities. As <strong>the</strong> largest and most<br />

diverse group <strong>of</strong> transmembrane proteins, GPCRs represent<br />

<strong>the</strong> most druggable <strong>target</strong>s. However, due to <strong>the</strong> difficulty in<br />

getting soluble and active proteins, GPCRs are difficult<br />

Copyright # 2005 John Wiley & Sons, Ltd.


328 L. HUANG ET AL.<br />

<strong>target</strong>s for any antibody-based technology. To conquer this<br />

difficulty, we developed a strategy to fully identify <strong>human</strong><br />

<strong>antibodies</strong> <strong>target</strong>ing <strong>C5aR</strong>.<br />

<strong>C5aR</strong> (CD88) is <strong>the</strong> receptor for C5a, a 74 amino acid<br />

peptide derived from <strong>the</strong> complement system following<br />

activation via <strong>the</strong> classical, alternative or lectin-binding<br />

pathways (Gerard and Gerard, 1994). C5a and its receptor<br />

<strong>C5aR</strong> mediate immunomodulatory and inflammatory activities<br />

such as chemotaxis, degranulation, vascular permeability<br />

changes and cytokine regulation, thus playing a central<br />

role in host defense <strong>against</strong> microorganisms and a diverse<br />

range <strong>of</strong> immunoinflammatory disorders (for a review, see<br />

Makrides, 1998; Pellas and Wennogle, 1999). <strong>C5aR</strong> is highly<br />

expressed on cells <strong>of</strong> myeloid origin including granulocytes,<br />

monocytes, lymphocytes and tissue inflammatory cells, including<br />

macrophages, microglia and mast cells. <strong>C5aR</strong> also is<br />

expressed, albeit at a lower level, on non-myeloid cells such<br />

as epi<strong>the</strong>lial, endo<strong>the</strong>lial and smooth muscle cells in <strong>the</strong> liver<br />

and lung. Owing to <strong>the</strong> high expression <strong>of</strong> <strong>C5aR</strong> on blood<br />

leukocytes and tissue inflammatory cells as well as <strong>the</strong><br />

central role that this ligand–receptor complex plays in a<br />

number <strong>of</strong> inflammatory disorders, <strong>C5aR</strong> and C5a have been<br />

<strong>target</strong>s for <strong>the</strong>rapeutic intervention. A large focus <strong>of</strong> this<br />

effort has been on <strong>the</strong> identification and development <strong>of</strong><br />

small molecule inhibitors to <strong>the</strong> receptor and neutralizing<br />

<strong>antibodies</strong> to C5a. Antibodies <strong>target</strong>ing <strong>C5aR</strong> will have <strong>the</strong><br />

potential to be developed as agents for <strong>the</strong>rapeutics and<br />

infection/inflammation imaging.<br />

With our Fab displaying phage library and rapid reformatting<br />

procedure, we were able to discover fully <strong>human</strong><br />

anti-<strong>C5aR</strong> <strong>antibodies</strong> within a short period <strong>of</strong> time, demonstrating<br />

that difficult <strong>target</strong>s such as GPCRs can be successfully<br />

<strong>target</strong>ed <strong>using</strong> this approach.<br />

EXPERIMENTAL PROCEDURES<br />

Cell culture<br />

The <strong>human</strong> embryonic kidney HEK293T cells (GenHunter,<br />

catalog no. Q401) were cultured in DMEM supplemented<br />

with 10% ‘ultralow IgG’ fetal calf serum (Invitrogen,<br />

catalog no. 31966021). The promonocytic U-937 cells<br />

(ATCC, catalog no. CRL-1593.2) were cultured in RPMI<br />

1640 medium supplemented with 10% heat-inactivated fetal<br />

calf serum (FCS), 2 mM L-glutamine, 0.1 mM non-essential<br />

amino acids, 1 mM sodium pyruvate, 100 Unit/ml <strong>of</strong> penicillin,<br />

and 100 mg/ml <strong>of</strong> streptomycin. To induce differentiation<br />

<strong>of</strong> U-937 cells, cells were treated with <strong>the</strong> membranepermeable<br />

cAMP analog, N 6 ,2 0 -o-dibutyryladenosine 3 0 ,5 0<br />

cyclic monophosphate (Bt 2 cAMP) (Sigma, catalog no.<br />

C5788) at 0.5 mM for 3 days. All cell cultures were incubated<br />

at 37 C in a humidified atmosphere with 5% CO 2 .<br />

Peptide syn<strong>the</strong>sis and labeling<br />

The <strong>C5aR</strong> N-terminal peptide (amino acid 1–31) with an<br />

addition <strong>of</strong> lysine residue at <strong>the</strong> C-terminus (NH2-<br />

MNSFNYTTPDYGHYDDKDTLDLNTPVDKTSNK-NH2)<br />

was syn<strong>the</strong>sized on solid phase <strong>using</strong> 9-fluorenylmethoxycarbonyl<br />

protocols. A portion <strong>of</strong> <strong>the</strong> syn<strong>the</strong>sized peptide was<br />

labeled on resin with <strong>the</strong> dPEG4/Biotin derivative via <strong>the</strong><br />

lysine residue. Both labeled and unlabeled peptides were<br />

cleaved from resin with trifluoroacetic acid, purified <strong>using</strong><br />

reversed-phase high-performance liquid chromatography,<br />

confirmed by electrospray mass spectrometry, and lyophilized.<br />

Peptide concentrations were determined by measuring<br />

<strong>the</strong> absorbance at 280 nm. The purity <strong>of</strong> each peptide was<br />

greater than 99.9%.<br />

Library construction<br />

The <strong>human</strong> Fab phage display library (Fab 400) was constructed<br />

as described (Hoet et al., 2004). Briefly, V-genes<br />

from autoimmune patients (V L C L and V H -CDR3) and<br />

syn<strong>the</strong>tic V H -CDR1/2 were cloned into a phage display<br />

vector, <strong>using</strong> a completely novel cloning method, oligonucleotide-assisted<br />

cleavage and ligation (ONCL). The library<br />

has a diversity <strong>of</strong> 1 10 10 .<br />

Library selection<br />

The biotinylated 31 amino acid <strong>C5aR</strong> N-terminal peptide<br />

(DX-1186) was used as <strong>the</strong> <strong>target</strong> for selection. Magnetic<br />

Dynabeads M-280 streptavidin (Dynal Biotech Inc., catalog<br />

no. 112.06) were used to immobilize DX-1186. In round 1,<br />

input phage (1 10 12 PFU <strong>of</strong> Fab400 library per selection)<br />

and beads were each blocked at room temperature (RT) for<br />

1 h with phosphate buffered saline (PBS) containing 2% (w/<br />

v) non-fat dry milk and 0.1% (v/v) Tween-20. Blocked<br />

phage were allowed to bind <strong>the</strong> free <strong>target</strong> DX-1186 in<br />

solution (liquid binding selection) or <strong>the</strong> <strong>target</strong> immobilized<br />

on beads (bead binding selection). In liquid binding selection,<br />

after incubation with free DX-1186 (1 mM) at RT for<br />

1 h, <strong>the</strong> phage <strong>target</strong> mixture was applied to beads for 15 min<br />

at RT. The beads were washed seven times with PBS<br />

containing 0.1% (v/v) Tween-20 (PBST) to remove unbound<br />

phage. The bound phage were <strong>the</strong>n eluted with<br />

10 mM non-biotinylated peptide DX-1185 in PBST by incubating<br />

at RT for 1 h. Eluted phage and remaining phage<br />

on beads were amplified by infecting E. coli cells, and<br />

underwent two more rounds <strong>of</strong> selection and amplification.<br />

The subsequent rounds <strong>of</strong> selection were essentially same as<br />

round 1, except depletion steps were included in rounds 2<br />

and 3 before incubating with <strong>the</strong> <strong>target</strong>. Depletion was<br />

carried out in rounds 2 and 3 to remove non-specific binders<br />

by incubating <strong>the</strong> input phage with beads for 15 min at RT on<br />

a rotator. The supernatant was removed from beads and<br />

subjected to two more depletion steps. The depleted input<br />

phage underwent selections as in round 1.<br />

Screening for <strong>C5aR</strong> peptide binders by phage ELISA<br />

Phage enriched from <strong>the</strong> third round <strong>of</strong> selection were<br />

screened by phage ELISA for <strong>C5aR</strong> peptide binding. Immulon<br />

2HB 96-well plates (ThermoLabsystems, catalog no.<br />

3455) were coated with 100 ng per well <strong>of</strong> streptavidin<br />

(Pierce, catalog no. 21120) diluted in PBS for 1 h at<br />

37 C. The <strong>target</strong> plates were fur<strong>the</strong>r coated with 40 ng<br />

per well <strong>of</strong> DX-1186 diluted in PBS for 1 h at RT. The<br />

Copyright # 2005 John Wiley & Sons, Ltd. J. Mol. Recognit. 2005; 18: 327–333


HUMAN ANTIBODIES AGAINST THE <strong>C5aR</strong> TARGET 329<br />

background plates were incubated with PBS. The coated<br />

plates were blocked with 1% (w/v) bovine serum albumin<br />

(BSA) in PBS for 1 h at 37 C, and washed five times with<br />

PBST. The plates were <strong>the</strong>n incubated for 1 h with 1:2<br />

diluted overnight phage cultures that were produced by<br />

inoculating phage from individual plaques into E. coli cells.<br />

After washing seven times with PBST, <strong>the</strong> plates were<br />

incubated with horseradish peroxidase (HRP)-conjugated<br />

anti-M13 antibody (Amersham Pharmacia, catalog no.<br />

27-9421-01) for 1 h, washed seven times, developed with<br />

tetramethylbenzidine (TMB) peroxidase substrate solution<br />

(Kirkegaard & Perry Laboratories, catalog no. 50-76-03)<br />

and read at 630 nm on an ELISA plate reader.<br />

Reformatting <strong>of</strong> Fabs into whole IgG <strong>antibodies</strong><br />

IgG reformatting was carried out as described (Jostock et al.,<br />

2004). Briefly, <strong>the</strong> Fab cassette containing a prokaryotic<br />

ribosome binding site (RBS) was lifted from <strong>the</strong> phage<br />

display vector by PCR and inserted into <strong>the</strong> IgG expression<br />

vector. The prokaryotic RBS was <strong>the</strong>n replaced with a<br />

eukaryotic internal ribosome entry site (IRES). To express<br />

IgG <strong>antibodies</strong>, HEK293T cells were transiently transfected<br />

with <strong>the</strong> reformatted IgG vectors <strong>using</strong> Lip<strong>of</strong>ectamine 2000<br />

reagent (Invitrogen, catalog no. 11668019). Conditioned<br />

media were harvested 72 and 144 h after transfection,<br />

pooled, and sterile-filtered. IgGs were purified <strong>using</strong> protein<br />

A beads (Amersham Biosciences, catalog no. 17-1279-01).<br />

The final eluted IgGs were dialyzed <strong>against</strong> PBS, and IgG<br />

concentrations were determined by measuring <strong>the</strong> absorbance<br />

at 280 nm.<br />

RT. After washing seven times with PBST, <strong>the</strong> plate was<br />

incubated with a secondary HRP-conjugated anti-<strong>human</strong><br />

IgG (Bethyl, catalog no. A80-104P-38) for 1 h at RT,<br />

washed seven times, developed with TMB solutions, and<br />

read at 630 nm on an ELISA plate reader.<br />

RESULTS<br />

Selection <strong>of</strong> <strong>C5aR</strong> binders<br />

The 31 amino acid N-terminal extracellular domain <strong>of</strong> <strong>C5aR</strong><br />

was syn<strong>the</strong>sized and biotinylated via <strong>the</strong> lysine residue<br />

added at <strong>the</strong> C-terminus [Fig. 1(A)]. This biotinylated<br />

peptide, DX-1186, was used as <strong>the</strong> <strong>target</strong> for selection <strong>of</strong><br />

<strong>C5aR</strong> binders from a Fab phage display library (Fab 400)<br />

which was constructed <strong>using</strong> <strong>the</strong> novel ONCL method and<br />

contained novel V-gene composition resulting from a combination<br />

<strong>of</strong> natural and syn<strong>the</strong>tic diversity (Hoet et al.,<br />

2004). Two arms <strong>of</strong> selection were carried out: liquid<br />

binding and bead binding selections. For <strong>the</strong> liquid binding<br />

Flow cytometric analysis<br />

U-937 cells were harvested and resuspended in cold staining<br />

buffer [PBS, 0.1% (w/v) sodium azide, 1% (w/v) BSA]. For<br />

indirect immun<strong>of</strong>luorescence staining, cells were incubated<br />

with individual reformatted IgG antibody or <strong>the</strong> anti-<strong>C5aR</strong><br />

mouse monoclonal antibody (mAb) S5/1 (Serotec, catalog<br />

no. MCA1283XZ) at 10 mg/ml at 4 C for 20 min, washed,<br />

and fur<strong>the</strong>r incubated at 4 C for 20 min with a secondary<br />

fluorescein isothiocyanate (FITC)-conjugated goat anti-<strong>human</strong><br />

IgG (Jackson ImmunoResearch Laboratories, catalog<br />

no. 109-096-098) or goat anti-mouse IgG (Jackson ImmunoResearch<br />

Laboratories, catalog no. 115-096-072). Stained<br />

cells were <strong>the</strong>n washed and resuspended in staining buffer,<br />

and analyzed on a FACScan flow cytometer (Becton<br />

Dickinson).<br />

Competition ELISA<br />

The <strong>target</strong> wells in an ELISA plate were sequentially coated<br />

with streptavidin and DX-1186 as described above for <strong>the</strong><br />

phage ELISA. The background wells were only coated with<br />

streptavidin. The plate was blocked with 1% (w/v) BSA at<br />

37 C for 1 h, and washed five times with PBST. Prior to<br />

incubation with individual reformatted IgG at 1 mg/ml<br />

for 1 h at RT, <strong>the</strong> <strong>target</strong> wells were pre-incubated with<br />

mAb S5/1 or mouse IgG at 10-fold molar excess for 1 h at<br />

Figure 1. Library selection <strong>against</strong> <strong>the</strong> <strong>C5aR</strong> N-terminal peptide<br />

DX-1186 <strong>target</strong>. (A) Amino acid sequences <strong>of</strong> <strong>the</strong> <strong>target</strong> peptide<br />

DX-1186 and <strong>the</strong> peptide DX-1185 for elution. A lysine residue<br />

was added at <strong>the</strong> C-terminus <strong>of</strong> <strong>the</strong> peptide for labeling with<br />

dPEG4-biotin for DX-1186 peptide. DX-1185 was <strong>the</strong> unlabeled<br />

peptide for elution during selection. (B) Fraction <strong>of</strong> input (FOI)<br />

recovered after each round <strong>of</strong> selection. Three rounds <strong>of</strong> selection<br />

from <strong>the</strong> Fab phage library (Fab400) were carried out. FOI<br />

from each round was calculated as <strong>the</strong> total amount <strong>of</strong> output<br />

phage divided by <strong>the</strong> total amount <strong>of</strong> input phage. FOIs from<br />

both selections (liquid binding and bead binding) are shown.<br />

Copyright # 2005 John Wiley & Sons, Ltd. J. Mol. Recognit. 2005; 18: 327–333


330 L. HUANG ET AL.<br />

Figure 2. Representative phage ELISA results. Phage enriched from <strong>the</strong> third round <strong>of</strong> selection<br />

were screened for positive binders by phage ELISA. The <strong>target</strong> wells were sequentially coated<br />

with streptavidin and biotinylated DX-1186 peptide, and <strong>the</strong> background wells were coated only<br />

with streptavidin. Plotted in <strong>the</strong> histograms is absorbance at 630 nm (A630) <strong>of</strong> individual phage<br />

clone. Solid bar: DX-1186 <strong>target</strong> signal; open bar: streptavidin background signal. Shown here are<br />

representative ELISA data from two selections, liquid binding selection (A) and bead binding<br />

selection (B).<br />

selections, input phage were first incubated with <strong>the</strong> <strong>target</strong><br />

DX-1186 in solution, and <strong>the</strong>n immobilized to streptavidincoated<br />

magnetic beads via DX-1186. In <strong>the</strong> bead binding<br />

selection campaigns, input phage were allowed to bind <strong>the</strong><br />

immobilized <strong>target</strong> DX-1186 on streptavidin-coated magnetic<br />

beads. After removal <strong>of</strong> unbound phage by washing<br />

extensively with PBST, <strong>the</strong> bound phage were eluted with<br />

DX-1185, <strong>the</strong> non-biotinylated <strong>C5aR</strong> peptide [Fig. 1(A)].<br />

Eluted phage and <strong>the</strong> remaining phage on beads were<br />

subsequently amplified, and subjected to fur<strong>the</strong>r rounds <strong>of</strong><br />

selection. In rounds 2 and 3, depletion steps were carried out<br />

to remove non-specific phage binders. After three rounds <strong>of</strong><br />

selection, <strong>the</strong> fraction <strong>of</strong> input (FOI), which was calculated<br />

as <strong>the</strong> total amount <strong>of</strong> output phage divided by <strong>the</strong> total<br />

amount <strong>of</strong> input phage, increased from 10 7 –10 6 at <strong>the</strong><br />

first round to 10 3 –10 2 by <strong>the</strong> third round [Fig. 1(B)],<br />

suggesting enrichment <strong>of</strong> phage binders to <strong>the</strong> <strong>target</strong>.<br />

To identify positive phage binders, <strong>the</strong> eluted phage from<br />

<strong>the</strong> third round <strong>of</strong> selection were screened by phage ELISA.<br />

The <strong>target</strong> plates were sequentially coated with streptavidin<br />

and DX-1186; and <strong>the</strong> background plates were coated only<br />

with streptavidin. As shown in Fig. 2 for representative<br />

ELISA data and Table 1 for summary <strong>of</strong> results, <strong>the</strong> hit rates<br />

<strong>of</strong> positive isolates with ELISA signals greater than 5-fold<br />

over background levels were high from both selection<br />

campaigns: 44% for liquid binding selection and 53% for<br />

bead binding selection. If phage isolates with ELISA signals<br />

greater than 3-fold over background levels were considered<br />

positive, <strong>the</strong>n <strong>the</strong> hit rates were more than 80% from both<br />

selections. As noted in Fig. 2, a significant number <strong>of</strong> phage<br />

isolates showed no to very low ELISA signals (A630 < 0.2).<br />

These phage isolates were ei<strong>the</strong>r Fab-displaying phage that<br />

did not bind <strong>the</strong> <strong>target</strong> or phage with no Fab display, and thus<br />

served as good internal negative controls for <strong>the</strong> assay.<br />

Therefore, <strong>the</strong> selection campaigns on peptide <strong>target</strong> were<br />

successful in identifying Fab-phage binders to <strong>the</strong> <strong>target</strong><br />

peptide.<br />

Table 1. Summary <strong>of</strong> phage ELISA results<br />

Selection arm Liquid binding Bead binding<br />

Total isolates 384 384<br />

Positive isolates a 170 204<br />

Hit rate (%) 44 53<br />

a Isolates with ELISA signals greater than 5-fold over streptavidin<br />

background levels.<br />

Copyright # 2005 John Wiley & Sons, Ltd. J. Mol. Recognit. 2005; 18: 327–333


HUMAN ANTIBODIES AGAINST THE <strong>C5aR</strong> TARGET 331<br />

Sequencing <strong>of</strong> Fab-phage isolates and IgG reformatting<br />

Selected ELISA positive isolates were subsequently sequenced.<br />

Sequence alignment analysis <strong>of</strong> Fab amino acid<br />

sequences <strong>of</strong> 77 isolates with full Fab sequence data available<br />

resulted in <strong>the</strong> classification <strong>of</strong> <strong>the</strong> ELISA hits into five<br />

major groups, based on heavy chain (HC) CDR1–3 usage.<br />

Each major group has <strong>the</strong> same HC CDRs, but different light<br />

chain (LC) CDRs. Variations <strong>of</strong> LC usage fur<strong>the</strong>r divided<br />

each group into subgroups, with a total number <strong>of</strong> groups<br />

being 22. Besides <strong>the</strong> 22 subgroups, <strong>the</strong>re were seven unique<br />

sequences. Representative phage clones, a total <strong>of</strong> 29 isolates,<br />

were chosen from each subgroup and unique isolates,<br />

and reformatted into whole IgG molecules. Sixteen<br />

Figure 3. FACS analysis <strong>of</strong> reformatted IgGs. U-937 cells were differentiated into macrophages by treatment with Bt 2 cAMP at<br />

0.5 mM for 3 days. Differentiated cells were stained with IgGs by indirect immun<strong>of</strong>luorescence. Cells were first incubated with<br />

reformatted IgGs and control <strong>human</strong> IgG (hIgG) in staining buffer. S5/1, which binds to <strong>the</strong> <strong>C5aR</strong> N-terminal region, was used<br />

as a positive control. Mouse IgG (mIgG) was used as an isotype control for S5/1. After washing, cells were stained with a FITClabeled<br />

secondary antibody, and <strong>the</strong>n analyzed by flow cytometry. (A) Individual histograms <strong>of</strong> control hIgG, selected<br />

reformatted IgGs (IgG-4, 32, 15, 28, 31, 33, 38, 43), control mIgG and S5/1. The x-axis depicts <strong>the</strong> fluorescence intensity <strong>of</strong><br />

individual cells, and <strong>the</strong> y-axis represents <strong>the</strong> cell number. (B) Summary <strong>of</strong> percentage positive cells for all IgGs tested. The y-<br />

axis represents <strong>the</strong> relative percentages <strong>of</strong> positively stained cells, which were set based on negative controls. Data shown<br />

are representative <strong>of</strong> three independent experiments.<br />

Copyright # 2005 John Wiley & Sons, Ltd. J. Mol. Recognit. 2005; 18: 327–333


332 L. HUANG ET AL.<br />

reformatted clones were expressed as IgGs (5–15 mg/ml) in<br />

transiently transfected HEK293T cells. IgGs from <strong>the</strong>se 16<br />

clones were subsequently purified, and tested in cell binding<br />

assays.<br />

Testing reformatted IgGs for cell binding activities<br />

Since syn<strong>the</strong>tic peptides lack post-translational modifications<br />

such as glycosylation and may adopt conformations different<br />

to <strong>the</strong> same sequence in <strong>the</strong> native protein, <strong>antibodies</strong><br />

discovered from selection on peptide <strong>target</strong>s need to be<br />

fur<strong>the</strong>r evaluated in cell-based assays. Reformatted IgGs<br />

were thus tested by flow cytometric analysis for <strong>the</strong>ir cell<br />

binding activities on <strong>C5aR</strong>-expressing cells. <strong>C5aR</strong> was<br />

induced in differentiated promonocytic U-937 cells (Gerard<br />

and Gerard, 1990). U-937 cells were terminally differentiated<br />

by treatment with 0.5 mM Bt 2 cAMP for 3 days, and <strong>the</strong>n<br />

stained with IgGs by indirect immun<strong>of</strong>luorescence. Cells<br />

were first incubated with reformatted IgGs and control <strong>human</strong><br />

IgG (hIgG) in staining buffer. S5/1, amAb that recognizes<br />

<strong>the</strong> <strong>C5aR</strong> N-terminal region (Oppermann et al., 1993)<br />

was used as a positive control. Flow cytometric analysis<br />

demonstrated that six reformatted IgGs (IgG-15, 28, 31, 33,<br />

38 and 43) showed good binding to differentiated U-937<br />

cells, with percentages <strong>of</strong> positive cells ranging from 11–30%<br />

(Fig. 3). The positive control antibody S5/1 also showed<br />

binding to cells with <strong>the</strong> percentage <strong>of</strong> positive cells being<br />

approximately 17%. The o<strong>the</strong>r 10 reformatted IgGs (IgG-4,<br />

11, 12, 16, 17, 18, 25, 29, 32 and 37) did not show significant<br />

binding to cells, indicating that <strong>the</strong>y were not cell binders.<br />

Consistent with <strong>the</strong> <strong>C5aR</strong> expression pr<strong>of</strong>ile that this<br />

GPCR was not significantly expressed on undifferentiated<br />

U-937 cells but was induced in differentiated U-937 cells,<br />

one representative positive cell binder IgG-28 showed<br />

binding in differentiated U-937 cells but not in undifferentiated<br />

cells (Fig. 4), suggesting that IgG-28 is specific to<br />

differentiated U-937 cells.<br />

Figure 5. Specificity <strong>of</strong> positive cell binders to <strong>C5aR</strong>. Competition<br />

ELISA was carried out as described in <strong>the</strong> Experimental<br />

Procedures. The <strong>target</strong> wells were sequentially coated with<br />

streptavidin and biotinylated DX-1186 peptide (white and striped<br />

bars), and <strong>the</strong> background wells were coated just with streptavidin<br />

(black bars). Prior to <strong>the</strong> addition <strong>of</strong> hIgG control and<br />

positive IgGs, <strong>target</strong> wells were incubated with 10 mIgG (white<br />

bars) or 10 S5/1 (striped bars). Plotted in <strong>the</strong> histograms are<br />

ELISA signals as measured by A630.<br />

Testing <strong>the</strong> specificities <strong>of</strong> positive cell binding IgGs<br />

To fur<strong>the</strong>r determine if <strong>the</strong> positive cell binding IgGs were<br />

specific to <strong>C5aR</strong>, <strong>the</strong> positive IgGs (IgG-15, 28, 31, 33, 38<br />

and 43) were tested in a peptide-based ELISA. Each <strong>of</strong> <strong>the</strong> 6<br />

IgGs bound to DX-1186 (Fig. 5). Competition ELISA with<br />

mAb S5/1 known to bind <strong>the</strong> same <strong>C5aR</strong> N-terminal region<br />

showed that all six IgGs were competed with by S5/1<br />

(Fig. 5), indicating that <strong>the</strong>se positive IgG cell binders<br />

were specific to <strong>C5aR</strong>. This was fur<strong>the</strong>r confirmed by<br />

FACS analysis <strong>of</strong> <strong>C5aR</strong>-expressing cells showing that <strong>the</strong><br />

IgG binding to cells was competed by S5/1 (data not<br />

shown).<br />

DISCUSSION<br />

Figure 4. Specific binding <strong>of</strong> positive IgG binders to differentiated<br />

U-937 cells. U-937 cells treated with Bt 2 cAMP at 0.5 mM for<br />

3 days were used as differentiated cells, and untreated cells were<br />

used as undifferentiated cells. Both differentiated and undifferentiated<br />

cells were stained by indirect immun<strong>of</strong>luorescence and<br />

analyzed by flow cytometry. Shown here are histograms <strong>of</strong><br />

representative positive IgG binder, IgG-28.<br />

We have used phage display technology to identify <strong>human</strong><br />

<strong>antibodies</strong> to <strong>the</strong> chemoattractant receptor <strong>C5aR</strong>, a member<br />

<strong>of</strong> <strong>the</strong> GPCR superfamily. GPCRs have proved successful<br />

drug <strong>target</strong>s and a large proportion <strong>of</strong> existing drugs <strong>target</strong><br />

<strong>the</strong>se receptors. However, owing to <strong>the</strong> complex structures<br />

<strong>of</strong> <strong>the</strong>se seven transmembrane domain-based proteins,<br />

GPCRs are <strong>of</strong>ten considered difficult <strong>target</strong>s for antibodybased<br />

<strong>the</strong>rapeutics and <strong>target</strong>ing. We have designed a<br />

strategy <strong>using</strong> a syn<strong>the</strong>tic peptide encompassing <strong>the</strong> N-<br />

terminal region <strong>of</strong> <strong>C5aR</strong> as <strong>the</strong> selection <strong>target</strong>. After three<br />

rounds <strong>of</strong> selection with a Fab phage library, we were able to<br />

obtain very high hit rates (50% <strong>of</strong> isolates with ELISA<br />

signals greater than 5-fold over background levels) utilizing<br />

two selection strategies: liquid binding and bead binding. In<br />

liquid binding selection, <strong>the</strong> input phage were incubated<br />

with <strong>the</strong> biotin-conjugated <strong>target</strong> in solution before capture<br />

Copyright # 2005 John Wiley & Sons, Ltd. J. Mol. Recognit. 2005; 18: 327–333


HUMAN ANTIBODIES AGAINST THE <strong>C5aR</strong> TARGET 333<br />

on streptavidin beads; in bead binding selection, <strong>the</strong> input<br />

phage were incubated with <strong>the</strong> <strong>target</strong> immobilized on beads.<br />

There are advantages and disadvantages for both selection<br />

strategies. In liquid binding selection, high-affinity binders<br />

may be enriched due to <strong>the</strong> lack <strong>of</strong> avidity effect, but <strong>the</strong><br />

<strong>target</strong> concentration should be well controlled so that <strong>the</strong><br />

<strong>target</strong> is well below <strong>the</strong> saturating concentration and potential<br />

binders will not be lost after immobilizing to beads. In<br />

bead binding selection, low-affinity binders also may be<br />

enriched due to avidity effect, but <strong>the</strong> <strong>target</strong> concentrations<br />

did not need to be well-controlled as in liquid binding<br />

selection. In this study, we included both selection strategies<br />

to ensure maximum success. Our results showed that both<br />

selections worked very well, resulting in similar hit rates.<br />

Representative Fab clones were reformatted into IgGs, and<br />

tested for cell binding activities by flow cytometry. Approximately<br />

40% <strong>of</strong> <strong>the</strong> reformatted IgGs showed binding to<br />

differentiated U-937 cells in which <strong>C5aR</strong> expression was<br />

induced. Flow cytometric analysis <strong>of</strong> HepG2 cells known to<br />

express <strong>C5aR</strong> (Haviland et al., 1995) paralleled <strong>the</strong> results<br />

from U-937 cells: positive binders to differentiated U-937<br />

cells were also positive binders to HepG2 cells, and negative<br />

binders to differentiated U-937 cells were also negative<br />

binders to HepG2 cells (data not shown). These positive<br />

cell binders were specific for <strong>C5aR</strong> since <strong>the</strong>y bound to<br />

<strong>C5aR</strong> N-terminal peptide DX-1186 in peptide-based<br />

ELISA, and <strong>the</strong>ir binding was competed by S5/1, an<br />

anti-<strong>C5aR</strong> mAb known to bind <strong>the</strong> same <strong>C5aR</strong> N-terminal<br />

region (Oppermann et al., 1993). The cell binding activities<br />

<strong>of</strong> <strong>the</strong>se positive binders also were competed by<br />

S5/1, suggesting specific binding to <strong>the</strong> <strong>C5aR</strong> N-terminal<br />

region.<br />

The library used here for selection is a Fab phage display<br />

library with combined natural and syn<strong>the</strong>tic diversity (Hoet<br />

et al., 2004). The advantage <strong>of</strong> <strong>using</strong> a Fab phage library for<br />

selection is that <strong>the</strong>re is no need to do phagemid rescue<br />

during selection and phage ELISA, thus greatly reducing <strong>the</strong><br />

amount <strong>of</strong> time for selection and screening. The <strong>human</strong> Fab<br />

library has been used for selection with many soluble<br />

protein <strong>target</strong>s, yielding <strong>target</strong>-specific <strong>human</strong> <strong>antibodies</strong><br />

with high affinity (Hoet et al., 2004). With <strong>the</strong> Fab library<br />

and <strong>the</strong> IgG reformatting procedure (Jostock et al., 2004),<br />

we were able to obtain specific <strong>human</strong> IgGs to a GPCR<br />

within a short period <strong>of</strong> time (8 weeks), demonstrating<br />

that our phage display approach has utility for rapid development<br />

<strong>of</strong> antibody <strong>the</strong>rapeutic and imaging agents to<br />

GPCRs.<br />

Acknowledgments<br />

We thank Greg Conley for peptide syn<strong>the</strong>sis and Gary Bassil for DNA<br />

sequencing. We also thank Albert Edge for helpful discussions and Clive<br />

Wood for critical reading <strong>of</strong> <strong>the</strong> manuscript.<br />

REFERENCES<br />

Azzazy HME, Highsmith WE Jr. 2002. Phage display technology:<br />

clinical applications and recent innovations. Clin. Biochem.<br />

35: 425–445.<br />

Gerard C, Gerard NP. 1994. C5a anaphylatoxin and its seven transmembrane-segment<br />

receptor. A. Rev. Immunol. 12: 775–808.<br />

Gerard NP, Gerard C. 1990. The chemotactic receptor for <strong>human</strong><br />

C5a anaphylatoxin. Nature 349: 614–617.<br />

Haviland DL, McCoy RL, Whitehead WT, Akama H, Molmenti EP,<br />

Brown A, Haviland JC, Parks WC, Perlmutter DH, Wetsel RA.<br />

1995. Cellular expression <strong>of</strong> <strong>the</strong> C5a anaphylatoxin receptor<br />

(<strong>C5aR</strong>): demonstration <strong>of</strong> <strong>C5aR</strong> on nonmyeloid cells <strong>of</strong> <strong>the</strong><br />

liver and lung. J. Immunol. 154: 1861–1869.<br />

Hoet RM, Cohen EH, Kent RB, Rookey K, Schoonbroodt S, Hogan<br />

S, Rem L, Frans N, Daukandt M, Pieters H, van Hegelsom R,<br />

Coolen-van Neer N, Nastri HG, Rondon IJ, Leeds JA, Hufton<br />

SE, Huang L, Kashin I, Devlin M, Kuang G, Steukers M,<br />

Viswanathan M, Nixon AE, Sexton DJ, Hoogenboom HR,<br />

Ladner RC. 2004. High affinity <strong>human</strong> <strong>antibodies</strong> from<br />

combined syn<strong>the</strong>tic and captured diversity. Nat. Biotechnol.<br />

in press.<br />

Jostock T, Vanhove M, Brepoels E, van Gool R, Daukandt M,<br />

Wehnert A, van Hegelsom R, Dransfield D, Sexton D, Devlin<br />

M, Ley A, Mullberg J. 2004. Rapid generation <strong>of</strong> functional<br />

<strong>human</strong> IgG <strong>antibodies</strong> derived from Fab-on-phage display<br />

libraries. J. Immunol. Meth. 289: 65–80.<br />

Makrides SC. 1998. Therapeutic inhibition <strong>of</strong> <strong>the</strong> complement<br />

system. Pharmac. Rev. 50: 59–87.<br />

Oppermann M, Raedt U, Hebell T, Schmidt B, Zimmermann B,<br />

Gotze O. 1993. Probing <strong>the</strong> <strong>human</strong> receptor for C5a anaphylatoxin<br />

with site-directed <strong>antibodies</strong>. Identification <strong>of</strong> a potential<br />

ligand binding site on <strong>the</strong> NH2-terminal domain. J.<br />

Immunol. 151: 3785–3794.<br />

Pellas TC, Wennogle LP. 1999. C5a receptor antagonists. Curr.<br />

Pharm. Des. 5: 737–755.<br />

Stockwin LH, Holmes S. 2003. Antibodies as <strong>the</strong>rapeutic agents:<br />

vive la renaissance! Exp. Opin. Biol. Ther. 3: 1133–1152.<br />

Copyright # 2005 John Wiley & Sons, Ltd. J. Mol. Recognit. 2005; 18: 327–333

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!