You are on page 1of 13

ARTHRITIS & RHEUMATISM

Vol. 48, No. 11, November 2003, pp 3253–3265


DOI 10.1002/art.11299
© 2003, American College of Rheumatology

Generation and Characterization of LymphoStat-B, a Human


Monoclonal Antibody That Antagonizes the Bioactivities of
B Lymphocyte Stimulator

Kevin P. Baker,1 Bryan M. Edwards,2 Sarah H. Main,2 Gil H. Choi,1 Ruth E. Wager,1
Wendy G. Halpern,1 Patrick B. Lappin,3 Todd Riccobene,1 Donara Abramian,1
Les Sekut,1 Bonnie Sturm,1 Carol Poortman,1 Ralph R. Minter,2 Claire L. Dobson,2
Elizabeth Williams,2 Sara Carmen,2 Rodger Smith,1 Viktor Roschke,1 David M. Hilbert,1
Tristan J. Vaughan,2 and Vivian R. Albert1

Objective. To identify and characterize a fully 3 receptors, TACI, BCMA, and BLyS receptor
human antibody directed against B lymphocyte stimu- 3/BAFF-R. LymphoStat-B potently inhibited BLyS-
lator (BLyS), a tumor necrosis factor–related cytokine induced proliferation of B cells in vitro, and ad-
that plays a critical role in the regulation of B cell ministration of LymphoStat-B to mice prevented
maturation and development. Elevated levels of BLyS human BLyS-induced increases in splenic B cell num-
have been implicated in the pathogenesis of auto- bers and IgA titers. In cynomolgus monkeys, adminis-
immune diseases. tration of LymphoStat-B resulted in decreased B cell
Methods. A human phage display library was representation in both spleen and mesenteric lymph
screened for antibodies against human BLyS. A human nodes.
monoclonal antibody, LymphoStat-B, specific for hu- Conclusion. A fully human monoclonal antibody
man BLyS was obtained from the library screening and has been isolated that binds to BLyS with high affinity
subsequent affinity optimization mutagenesis. The an- and neutralizes human BLyS bioactivity in vitro and in
tibody was tested for inhibition of human BLyS in vitro vivo. Administration of this antibody to cynomolgus
and in an in vivo murine model. Additionally, the monkeys resulted in B cell depletion in spleen and
consequences of BLyS inhibition were tested in vivo by lymph node. This antibody may prove therapeutically
administration of LymphoStat-B to cynomolgus mon- useful in the treatment of autoimmune diseases in
keys. humans.
Results. LymphoStat-B bound with high affinity
to human BLyS and inhibited the binding of BLyS to its
B lymphocyte stimulator (BLyS; trademark of
1
Kevin P. Baker, PhD, Gil H. Choi, PhD, Ruth E. Wager, Human Genome Sciences, Rockville, MD) protein (also
PhD, Wendy G. Halpern, DVM, PhD, Todd Riccobene, PhD, Donara known as BAFF, THANK, TALL-1, TNFSF13B, and
Abramian, MS, Les Sekut, DVM, Bonnie Sturm, BS, Carol Poortman,
BS, Rodger Smith, PhD, Viktor Roschke, PhD, David M. Hilbert, zTNF4), a member of the tumor necrosis factor (TNF)
PhD, Vivian R. Albert, PhD: Human Genome Sciences, Inc., Rock- ligand superfamily, is synthesized as a 285–amino acid
ville, Maryland; 2Bryan M. Edwards, PhD, Sarah H. Main, MSc, Ralph type II membrane protein and exists in both membrane
R. Minter, PhD, Claire L. Dobson, PhD, Elizabeth Williams, BSc, Sara
Carmen, BSc, Tristan J. Vaughan, PhD: Cambridge Antibody Tech- and cleaved 152–amino acid soluble forms (1–6). BLyS
nology, Granta Park, Cambridge, UK; 3Patrick B. Lappin, DVM, PhD: is expressed on monocytes, macrophages, and monocyte-
Charles River Laboratories, Sierra Biomedical Division, Sparks, Ne- derived dendritic cells, and is up-regulated in response
vada.
Address correspondence and reprint requests to Kevin P. to interferon-␥ and interleukin-10 (IL-10) (7). In vitro,
Baker, PhD, Department of Antibody Discovery and Development, recombinant human BLyS enhances B cell proliferation
Human Genome Sciences, Inc., 9410 Key West Avenue, Rockville, and Ig secretion through interaction with receptors
MD 20850. E-mail: kevin_baker@hgsi.com.
Submitted for publication March 5, 2003; accepted in revised expressed predominantly on B cells (1). In vivo, recom-
form July 3, 2003. binant human BLyS causes splenic hyperplasia in mice,
3253
3254 BAKER ET AL

primarily due to increases in the number of mature B nists have shown efficacy in reducing disease manifesta-
cells. BLyS administration to mice also causes increases tions in murine models of both SLE and RA (6,27,28).
in the total serum Ig concentration and enhanced With the aim of developing a therapeutic agent for
humoral responses to both T cell–independent and T autoimmune disease, LymphoStat-B (Human Genome
cell–dependent antigens (1,2,8). Sciences) antibody, a human, neutralizing monoclonal
BLyS has been shown to bind with high affinity to antibody against human BLyS, was generated. We de-
3 receptors, all of which are members of the TNF scribe herein the generation of human antibodies against
receptor family (6,9,10). Two of the receptors, BCMA BLyS using phage display, as well as the in vitro and in
and TACI, also bind APRIL, another TNF family mem- vivo characterization of LymphoStat-B.
ber that is the most homologous to BLyS (11–13). The
precise function of these two receptors is not well MATERIALS AND METHODS
understood, but studies in knockout mice suggest that
BCMA is functionally redundant, since BCMA-deficient Phage display libraries and selections. Nonimmunized
human single-chain variable fragment (scFv) phage display
mice have a normal B cell phenotype (14). TACI has libraries, recently expanded from 1010–1011 clones (29), were
been shown to have an inhibitory role in B cell develop- used for lead-scFv isolation. BLyS, at 10 ␮g/ml in phosphate
ment, since TACI knockout animals exhibit increased buffered saline (PBS), was immobilized on immunotubes
peripheral B cells, reduced responses to T cell– (Nunc, Naperville, IL), and BLyS-binding phage was isolated
independent antigens (15,16), and lymphoproliferative by 3 sequential rounds of panning (29). The optimized variants
were isolated by selection from randomized libraries in solu-
disorders and autoimmune disease (17). In contrast, tion using biotinylated BLyS captured on streptavidin-coated
BLyS-deficient mice show a phenotype of severe loss of paramagnetic beads (Dynal, New Hyde Park, NY) (30). The
mature B cells in the spleen, peripheral blood, and hBLySsc-1 and hBLySsc-2 V H third complementarity-
lymph nodes (18,19). A third receptor, BLyS receptor 3 determining region (CDR3) randomized repertoires were con-
(BR-3; BAFF-R), is specific for BLyS (9,10). The structed by polymerase chain reaction (PCR) using mutagenic
oligonucleotides to replace the last 6 VH CDR3 amino acids
A/WySnJ mouse strain, which harbors a naturally occur- with randomized codons. The mutated DNA for both lineages
ring truncation of BR-3, exhibits a phenotype similar to was ligated into the phagemid vector pCANTAB6 (31) and
that of BLyS-deficient mice (9,10,20), suggesting that electroporated into Escherichia coli TG1 (32). Libraries of 2 ⫻
BR-3 is the primary mediator of the effects of BLyS on 108 and 6 ⫻ 108 individual clones were generated for
B cell survival and maturation. hBLySsc-1 and hBLySsc-2, respectively. The randomized li-
braries were subjected to 1 round of panning on 10 ␮g/ml of
Several lines of evidence suggest that elevated immobilized BLyS, followed by 12 rounds of soluble selection
levels of BLyS may be involved in the pathogenesis of B using decreasing concentrations of biotinylated BLyS, from 50
cell–mediated autoimmune diseases. First, constitutive nM down to 100 pM.
overexpression of BLyS in transgenic animals results in Preparation and screening of scFv in a receptor bind-
manifestations of autoimmune-like symptoms, including ing inhibition assay. Expression of scFv was induced in 2TY
medium (1.6% weight/volume tryptone, 1% w/v yeast extract,
anti-DNA antibodies, rheumatoid factor, circulating im- 0.5% w/v NaCl) containing 1 mM IPTG for 3 hours at 30°C.
mune complexes, and deposition of immune complexes Periplasmic extracts (33) were prepared, and His-tagged scFv
in the kidney leading to glomerulonephritis. These were purified on nickel agarose (Qiagen, Chatsworth, CA) and
symptoms resemble those of systemic lupus erythemato- buffer exchanged into PBS. BLyS was biotinylated using
sus (SLE) and some aspects of rheumatoid arthritis N-hydroxysuccinimide (NHS)–biotin (Pierce, Rockford, IL) at
a molar ratio of 20:1 of biotin to BLyS. IM9 cells were
(RA) (21,22). Second, elevated levels of BLyS have been immobilized onto poly-L-lysine–coated 96-well plates at a
found in other murine models of SLE, including MRL- density of 1 ⫻ 105 cells/well, and scFv was added in the
lpr/lpr and (NZB ⫻ NZW)F1 strains (6). Finally, ele- presence of 25 ng/ml (0.5 nM) of biotinylated BLyS. Bound
vated levels of BLyS have been observed in the serum of biotinylated BLyS was detected via streptavidin–Delfia (Perkin
patients with SLE and RA (23,24) as well as Sjögren’s Elmer, Boston, MA). The 50% inhibition concentration (IC50)
values for the competition of each scFv for BLyS binding to its
syndrome (25,26). Significant correlations between receptor were then determined.
BLyS levels and autoantibody production were shown in Cloning and expression of antibodies. Variable heavy
these studies. and light chain fragments from scFv clones were subcloned as
The association of BLyS overproduction with separate heavy and light chains into expression vectors ob-
manifestations of several autoimmune diseases suggests tained from Cambridge Antibody Technology (34). Expression
constructs were used for transfection of mammalian cell lines
that modulation of BLyS levels could be a novel thera- (HEK293T or NSO) to produce whole IgG. Conditioned
peutic approach to the treatment of such diseases. media were collected 72 hours posttransfection, and antibodies
Indeed, studies with soluble BLyS receptors as antago- were purified by protein A chromatography.
LYMPHOSTAT-B, HUMAN MONOCLONAL ANTIBODY AGAINST BLyS 3255

Binding to BLyS in solid phase (direct enzyme-linked NaCl, pH 6) was injected subcutaneously 1 hour after antibody
immunosorbent assay [ELISA]). ELISA plates (Immulon-II; injection. On day 3, mice received a second dose of antibody (1
Dynatech, Chantilly, VA) were coated with 1 ␮g/ml of strepta- hour before BLyS injection). BLyS was injected subcutane-
vidin (Sigma, St. Louis, MO) in PBS. After washing in PBS and ously once a day for 4 consecutive days. On day 5, all mice were
blocking with 3% bovine serum albumin (BSA), 100 ␮l of killed 24 hours after the last BLyS injection. Sera were
biotinylated BLyS solution in PBST (1 ␮g/ml in PBS, 0.1% analyzed for IgA content by ELISA, wet spleen weights were
Tween 20, 0.1% BSA) was added to each well, and the wells recorded, and splenocytes were analyzed by flow cytometry
were incubated for 1 hour at room temperature. After washing, using phycoerythrin (PE)–conjugated anti–B220 (anti-
100 ␮l of serial dilutions of antibodies (66–6.6 ⫻ 10–6 nM) in CD45R) and fluorescein isothiocyanate (FITC)–conjugated
diluent buffer (PBS, 0.1% Tween 20, 0.1% BSA) was dis- anti-ThB (anti-Ly6D) as markers, as previously described
pensed into individual wells of BLyS-coated plates. The plates (1,35).
were incubated for 2 hours at room temperature, washed 4 Effects of LymphoStat-B in cynomolgus monkeys.
times with PBST, and then 100 ␮l of peroxidase-conjugated Studies with cynomolgus monkeys (Macaca fascicularis) were
goat anti-human IgG (1 ␮g/ml in diluent buffer; Vector, performed at Charles River Laboratories, Sierra Biomedical
Burlingame, CA) was added to each well. Plates were incu- Division (Sparks, NV). Monkeys (n ⫽ 10 [5 males and 5
bated for 1 hour at room temperature, washed 4 times with females] per dosage group) were given an intravenous injec-
PBST, and developed with tetramethylbenzidine substrate tion of vehicle or LymphoStat-B at 5, 15, or 50 mg/kg once a
(Sigma). Absorption at 450 nm was measured with a Spectra- week for 4 weeks. On day 29 (4 weeks of treatment), 3 monkeys
Max 3000 instrument (Molecular Devices, Sunnyvale, CA). of each sex from each dosage group were necropsied. The
Binding to BLyS in solution (competition ELISA). remaining monkeys were necropsied on day 57, after an
Serial dilutions of BLyS (103–10–4 nM) were prepared in additional 4-week treatment-free period. Periodically during
diluent buffer, and 50-␮l aliquots of each dilution were distrib- the study, the mononuclear cell populations in peripheral
uted to individual wells of BLyS-coated plates (prepared as blood were evaluated by flow cytometry using a Coulter Epics
described above). Antibodies were diluted to the 50% maxi- XL-MCL instrument (Beckman-Coulter, Miami, FL). In addi-
mum response concentration (EC50), and 50 ␮l of the resultant tion, relative mononuclear cell populations in the spleen and
dilution was added to the wells already containing BLyS. The mesenteric lymph nodes were determined for each monkey at
plates were further processed as described above. the time of necropsy. For both peripheral blood and tissues,
Murine splenocyte in vitro proliferation assay. Serial the following markers were used to distinguish immunopheno-
3-fold antibody dilutions were prepared in a 96-well plate. The type: CD2 for total lymphocytes, CD20 for B cells, CD20 and
final antibody concentrations ranged from 100 nM to 0.01 nM; CD21 for mature B cells, CD3 for total T cells, CD3 and CD4
a preparation containing no antibody served as a medium for helper T cells, CD3 and CD8 for suppressor/cytotoxic T
control. BLyS (3 ng/ml final concentration) that had been cells, and CD14 (CD3–) for monocytes. The PE-labeled
diluted in complete medium (RPMI 1640 with 10% fetal anti-CD2 antibody was obtained from Beckman-Coulter.
bovine serum containing 100 units/ml of penicillin, 100 ␮g/ml FITC-labeled anti-CD3 and anti-CD20 antibodies, as well
of streptomycin, 4 mM L -glutamine, and 5 ⫻ 10 –5 M as PE-labeled anti-CD4, anti-CD8, anti-CD14, and
␤-mercaptoethanol) was added to each well. Medium alone anti-CD21 antibodies, were obtained from BD PharMingen
(without BLyS) was used as the negative control for the assay (San Diego, CA).
background. Human IgG1 isotype controls were used as Inhibition of BLyS binding to BLyS receptor proteins.
negative controls for the antibodies. BLyS was labeled with the electrochemiluminescent reporter
Plates were incubated for 30 minutes at 37°C in an ORI-TAG–NHS (IGEN International, Gaithersburg, MD),
atmosphere containing 5% CO2. Splenocytes (final concentra- and binding to each of the 3 known BLyS receptors was
tion 1 ⫻ 106/ml) containing Staphylococcus aureus Cowan measured in the presence of LymphoStat-B, human IgG1
strain (Calbiochem, La Jolla, CA) were added to the wells. The isotype control antibody, free receptor fusion protein, or
plates were further incubated for 72 hours at 37°C in an control fusion protein herpesvirus entry mediator ([HVEM]–
atmosphere containing 5% CO2. Each well was supplemented Fc) essentially as previously described (36). Briefly, fusion
with complete medium containing 0.5 mCi of 3H-thymidine proteins of the extracellular domain of each of the 3 BLyS
(6.7 Ci/mmole; Amersham, Arlington Heights, IL), and the receptors (TACI, BCMA, and BR-3) and HVEM were biotin-
cells were incubated for an additional 20–24 hours at 37°C. The ylated with NHS–LC–biotin (Pierce) at molar ratios ranging
proliferation of each sample was measured by 3H-thymidine from 9:1 to 6.6:1. Twelve 2-fold serial dilutions of
incorporation and graphed as a function of antibody concen- LymphoStat-B, receptor, or controls at final concentrations
tration. A 4-parameter regression analysis was performed for ranging from 53 nM to 0.006 nM were prepared in assay
each data series, and IC50 values were derived. diluent, and 50-␮l aliquots of each dilution were added to
Neutralizing activity of LymphoStat-B antibody in individual wells of a 96-well plate. Fifty microliters of ORI-
mice. All animal experimentation was done in accordance with TAG–labeled BLyS at a final concentration of 40 ng/ml in
the Guide for the Care and Use of Laboratory Animals and assay diluent was added to each well, and the plate was
under the supervision of the Institutional Animal Care and incubated for 60–90 minutes at room temperature, with gentle
Use Committee. On day 1, female BALB/c mice (ages 8–9 shaking.
weeks) were injected intravenously with diluent (PBS) or with The biotinylated receptor proteins were each sepa-
LymphoStat-B or control human monoclonal IgG1 antibody at rately diluted to 50 ng/ml in assay diluent, and Dynabeads
doses of 0.05, 0.15, 0.5, 1.5, or 5 mg/kg. Recombinant human M280–streptavidin (Dynal) were used at a final concentration
BLyS at 0.3 mg/kg or BLyS vehicle (12.5 mM citrate, 125 mM of 250 ␮g/ml. One hundred microliters of this mixture was
3256 BAKER ET AL

BLyS either alone or in the presence of a 5-fold molar


excess of LymphoStat-B or an isotype control antibody.
BLyS binding was assessed with streptavidin–PE conjugate and
analyzed on a FACScan instrument (Becton Dickinson,
Mountain View, CA).
Flow cytometry analysis of LymphoStat-B binding.
Peripheral blood mononuclear cells were isolated from whole
human blood using lymphocyte separation medium (ICN
Biochemicals, Irvine, CA). Cells (10 6 in 100 ␮ l) in
fluorescence-activated cell sorting (FACS) buffer (PBS with
0.1% sodium azide and 0.1% BSA) were dispensed to FACS
tubes. Human IgG1 (20 ␮g in 10 ␮l) was added to each tube,
and the tubes were incubated for 20 minutes at 4°C to block
nonspecific Fc receptor–mediated binding. Biotinylated anti-
body or isotype control (1–2 ␮g) was added to each tube along
with FITC-conjugated lineage-specific marker antibody. Anti-

Figure 1. Identification of neutralizing antibodies to human B lym-


phocyte stimulator (BLyS). A, Purified BLyS-specific single-chain
variable fragments (scFv) were evaluated for the ability to inhibit
binding of biotinylated BLyS to its receptor on IM9 cells. The 50%
inhibition concentration (IC50) values are as follows: for hBLySsc-5,
14.12 nM; for hBLySsc-4, 18.23 nM; for hBLySsc-3, 20.63 nM; for
hBLySsc-2, 13.18 nM; and for hBLySsc-1, 2.69 nM; hBLySsc-6 did not
inhibit BLyS binding. B, Converted full IgG were evaluated for their
ability to neutralize BLyS-induced murine splenocyte proliferation as
measured by 3H-thymidine incorporation. The IC50 values are as
follows: for hBLySmAb-5, 0.09 nM; for hBLySmAb-4, 0.16 nM; for
hBLySmAb-3, 0.14 nM; for hBLySmAb-2, 0.08 nM; and for
hBLySmAb-1, 0.05 nM. A 4-parameter logistic model was used for
curve fitting and calculation of binding parameters. Values are the
mean ⫾ SEM of triplicate samples.

added to each well of the assay plate, and the plate was Figure 2. Improved potency of optimized BLyS single-chain antibod-
incubated for 45 minutes at room temperature, with vigorous ies in the receptor inhibition assay. Purified scFv were evaluated for
shaking. The plate was read on an Origen M8 series electro- their ability to inhibit biotinylated BLyS binding to its receptors on
chemiluminescence (ECL) analyzer (IGEN International), IM9 cells, as measured by europium-labeled streptavidin. A, Compar-
and the resulting ECL signal was plotted against the log ison of hBLySsc-1 and hBLySsc-1.1 (LymphoStat-B), showing that
nanomolar antibody or receptor concentration. hBLySsc-1.1 results in a 10-fold improvement in potency compared
Inhibition of receptor binding in a cellular system with the parental scFv hBLySsc-1. B, Comparison of hBLySsc-2 and
(flow cytometry). Flow cytometry was used to assess the hBLySsc-2.1, showing that hBLySsc-2.1 results in a 20-fold improve-
capacity of LymphoStat-B to inhibit binding of BLyS to ment in potency compared with the parental scFv hBLySsc-2. The IC50
HEK293T cells transfected with complementary DNA en- values are as follows: for hBLySsc-1, 6.3 nM; for hBLySsc-1.1, 0.5 nM;
coding full-length BLyS receptors. Cells were transfected with for hBLySsc-2, 5.86 nM; and for hBLySsc-2.1, 0.33 nM. A 4-parameter
TACI-, BCMA-, or BR-3–encoding plasmids and used for logistic model was used for curve fitting and calculation of binding
binding studies 24 hours posttransfection, as previously parameters. Values are the mean ⫾ SEM of triplicate samples. See
described (12). Cells were stained with 1 ␮g/ml of biotinylated Figure 1 for definitions.
LYMPHOSTAT-B, HUMAN MONOCLONAL ANTIBODY AGAINST BLyS 3257

demonstrated to specifically recognize BLyS and did not


bind to other TNF ligand family members, including
APRIL, LIGHT, TNF␣, Fas ligand, TRAIL, and TNF␤,
or to IL-4 or IL-18 (data not shown).
Optimization of lead scFv. Optimization of the
scFv corresponding to the lead candidates hBLySmAb-1
and hBLySmAb-2 was performed to identify antibodies
with enhanced inhibitory activity. Randomized libraries
were generated from the hBLySsc-1 and hBLySsc-2
sequences in which the terminal 6 residues of the VH
CDR3 were randomly replaced in order to increase
diversity within the VH domain. Optimized scFv were
again selected for binding to human BLyS.
The ability of the selected scFv to inhibit binding
of biotinylated BLyS to its receptors on the surface of
IM9 cells was assessed, and ⬎30 scFv were identified
with improved inhibitory profiles. Figure 2 illustrates the
Figure 3. Improved activity of optimized BLyS IgG in the murine
splenocyte proliferation assay. Converted full IgG molecules were
evaluated for their ability to inhibit BLyS-induced murine splenocyte
proliferation, as measured by 3H-thymidine incorporation. The IC50
values are as follows: for LymphoStat-B, 0.02 nM; for hBLySmAb-1,
0.05 nM, hBLySmAb-2.1, 0.05 nM; and for hBLySmAb-2, 0.08 nM. A
4-parameter logistic model was used for curve fitting and calculation of
binding parameters. Values are the mean ⫾ SEM of triplicate samples.
See Figure 1 for definitions.

body binding was assessed with streptavidin–PE (Dako,


Carpinteria, CA) measured in a FACScan instrument. The cell
line K562 was used as a positive control for the presence of
membrane-bound BLyS.

RESULTS
Isolation of BLyS binding antibodies. Large non-
immunized repertoires of human scFv fragments dis-
played on phage (29) were used for selection of single-
chain antibodies that bound to human BLyS protein.
More than 1,200 distinct BLyS-binding scFv were iden-
tified and were ranked by their ability to inhibit BLyS
binding to receptors on IM9 cells, a myeloma cell line
expressing significant levels of BLyS receptors. A panel
of 50 candidate scFv demonstrating the greatest inhibi-
tory activity was selected for further analysis. Inhibition
of binding activity for 6 of these scFv is shown in Figure
1A. These scFv were converted to full IgG molecules Figure 4. Binding of LymphoStat-B to B lymphocyte stimulator
and assessed for their ability to neutralize the activity of (BLyS), as determined by enzyme-linked immunosorbent assay. A,
human BLyS protein in a murine splenocyte in vitro Direct binding of LymphoStat-B to immobilized BLyS. B, Inhibition of
proliferation assay. Examples of antibodies demonstrat- LymphoStat-B binding to immobilized BLyS by BLyS in solution. The
calculated values for the 50% maximum response concentration and
ing the greatest neutralizing activity in this assay as full
the 50% inhibition concentration are 0.02 ⫾ 0.001 nM and 8.53 ⫾
IgG are shown in Figure 1B. Antibodies demonstrating 1.015 nM (mean ⫾ SEM of triplicate cultures), respectively. A
the best inhibitory profile as full IgG molecules were 4-parameter logistic model was used for curve fitting and calculation of
hBLySmAb-1 and hBLySmAb-2. Both antibodies were binding parameters. OD ⫽ optical density.
3258 BAKER ET AL

Table 1. Analysis of LymphoStat-B binding to membrane-bound Specificity of LymphoStat-B for soluble BLyS. In
BLyS* studies using a series of monoclonal antibodies against
Mean fluorescence intensity BLyS, differential recognition of membrane-bound
Human Mouse
BLyS has previously been reported (7), although the
IgG1 IgG1 biologic basis for this remains unclear. The ability of
Cell type control† LymphoStat-B control‡ 12D6 LymphoStat-B to recognize membrane-bound BLyS on
K562 cell line 7.19 5.26 3.48 6.34 the surface of cells was examined by flow cytometry
T lymphocytes (CD3⫹) 1.78 1.76 2.24 1.99 using primary human cells as well as the K562 myelog-
B lymphocytes (CD20⫹) 1.60 1.67 1.95 3.62 enous leukemia cell line that was previously character-
Natural killer cells 3.35 3.31 3.06 3.48
(CD56⫹) ized as expressing membrane-bound BLyS (Table 1).
Monocytes (CD14⫹) 4.06 3.29 2.93 9.20
Monocytes (CD14⫹) 8.46 5.13 3.87 21.55
plus interferon-␥

* Two-color fluorescence-activated cell sorter analysis was performed


on human peripheral blood mononuclear cells (PBMCs) or the cell
line K562 (positive control for the presence of membrane-bound B
lymphocyte) using biotinylated LymphoStat-B and fluorescein
isothiocyanate–labeled lineage-specific antibodies. PBMCs from 3
different donors were studied. A representative binding profile for 1
donor is presented. In all 3 donors, the 4 subpopulations of PBMCs
were negative for LymphoStat-B binding. In 2 of the 3 donors, the
murine monoclonal antibody detected BLyS on CD14⫹ monocytes
and possibly on a minor subpopulation of CD20⫹ cells.
† Negative control for LymphoStat-B binding.
‡ Negative control for 12D6 binding.

improvements that were observed for the 2 most potent


scFv from each lineage, hBLySsc-1.1 and hBLySsc-2.1,
compared with their respective parents. The hBLySsc-
1.1 isolate (IC50 0.7 nM) showed ⬃10-fold improvement
over hBLySsc-1 (Figure 2A), and hBLySsc-2.1 (IC50 0.2
nM) was improved ⬃20-fold over hBLySsc-2 (Figure
2B).
The top 30 scFv candidates were converted to full
IgG molecules and assayed for neutralization activity in
the murine splenocyte proliferation assay. Most full IgG
showed increased activity over the scFv form. The most
potent antibody in the neutralization assay, hBLySmAb-
1.1, was designated LymphoStat-B, and selected for
further characterization (Figure 3). The hBLySmAb-2.1
antibody, which showed the greatest inhibitory activity
as an scFv, did not demonstrate the greatest activity as a Figure 5. LymphoStat-B inhibition of B lymphocyte stimulator
full IgG. (BLyS) binding to A, TACI, B, BCMA, and C, BLyS receptor 3 (BR-3).
In vitro characterization of LymphoStat-B. Bind- LymphoStat-B was evaluated for its ability to inhibit the binding of
BLyS to extracellular domain fusion proteins of the 3 known BLyS
ing characteristics of LymphoStat-B were determined
receptors using an ORI-TAG–based electrochemiluminescence assay.
using a BLyS-specific ELISA in which biotinylated BLyS Binding is shown as electrochemiluminescence (ECL) counts. The
was immobilized in wells coated with streptavidin. As calculated 50% inhibition concentration (IC50) for LymphoStat-B
shown in Figure 4, LymphoStat-B bound to BLyS with inhibition of BLyS binding to TACI, BCMA, and BR-3 was 0.11, 0.10,
an EC50 value of 0.024 nM (Figure 4A). The binding of and 0.11 nM, respectively. The calculated IC50 value for each free
receptor fusion protein in the corresponding receptor-binding assay
LymphoStat-B was readily inhibited with soluble BLyS,
was 0.395 nM (TACI), 0.74 nM (BCMA), and 0.39 nM (BR-3). A
with an IC50 value of 8.5 nM (Figure 4B). Thus, 4-parameter logistic model was used for curve fitting and calculation of
LymphoStat-B binds BLyS in both a solid-phase capture binding parameters. Values are the mean ⫾ SEM of triplicate samples.
assay and in solution. Ab ⫽ antibody.
LYMPHOSTAT-B, HUMAN MONOCLONAL ANTIBODY AGAINST BLyS 3259

Figure 6. LymphoStat-B inhibition of B lymphocyte stimulator (BLyS) binding to full-length


TACI, BCMA, and BLyS receptor 3 (BR-3) expressed in HEK293T cells. Plasmids encoding the 3
full-length BLyS receptors or vector control were transfected into HEK293T cells. Cells were
subsequently stained with biotinylated BLyS, and binding was detected using a streptavidin–
phycoerythrin (PE) conjugate. Where indicated, a 5-fold molar excess (over BLyS) of
LymphoStat-B or human IgG1 control antibody was added to the staining reaction. Profiles marked
“No BLyS” represent staining of the streptavidin–PE conjugate alone (represented as the broken
line in the other plots).

The presence of membrane-bound BLyS was demon- it was important to assess the ability of LymphoStat-B to
strated by staining with a murine monoclonal antibody, inhibit BLyS binding to each receptor. Accordingly, the
12D6, which was previously characterized as recognizing extracellular domains of TACI, BCMA, and BR-3 were
membrane-bound BLyS. LymphoStat-B failed to stain expressed as fusion proteins, and the ability of
cells that were positive for monoclonal antibody 12D6. LymphoStat-B to inhibit labeled BLyS binding to each
Furthermore, interferon-␥, which has previously been was evaluated in an electrochemiluminescence detection
demonstrated to enhance BLyS expression on mono- assay. As shown in Figures 5A–C, LymphoStat-B inhib-
cytes, led to increased specific binding of the 12D6 ited BLyS binding to all 3 receptors with equivalent
antibody but not LymphoStat-B. These results suggest potency (IC50 0.10–0.11 nM).
that LymphoStat-B does not recognize membrane- The ability of LymphoStat-B to inhibit ligand–
bound BLyS. receptor interactions was also evident in a cell-based
LymphoStat-B inhibition of BLyS binding to assay in which full-length TACI, BCMA, and BR-3 were
TACI, BCMA, and BR-3. Recent studies have deter- transiently expressed on the surface of HEK293T cells
mined that BLyS binds to 3 cellular receptors, TACI, (Figure 6). BLyS-specific binding was readily detected
BCMA, and BR-3. Because the role of these receptors in on all receptor-expressing cells but not on cells trans-
B cell development and function is not fully understood, fected with the corresponding vector control. Addition
3260 BAKER ET AL

Figure 7. Inhibitory effects of LymphoStat-B on the responses of BALB/c mice stimulated with human B
lymphocyte stimulator (BLyS). BLyS was administered to mice over a 5-day period, with or without
LymphoStat-B or control IgG1 on days 1 and 3. The effects of BLyS on spleen weight, splenic B cell
representation, and serum IgA levels were determined on day 5. A, Effect of LymphoStat-B on
BLyS-induced increases in spleen weight. B, Effect of LymphoStat-B on BLyS-induced increases in the
number of mature splenic B cells (ThB⫹/B220⫹). These markers are the murine equivalents of Ly6D and
CD45R, respectively. Data are reported as the mean of the B220⫹/ThB⫹ cell population, as determined
by flow cytometry analysis. C, Effect of LymphoStat-B on BLyS-induced elevations of serum IgA levels.
Values are the mean ⫾ SEM (n ⫽ 10 mice per treatment group). ### ⫽ P ⬍ 0.0005 for recombinant
human BLyS versus buffer; ⴱ ⫽ P ⬍ 0.05 for LymphoStat-B versus the corresponding dose of human IgG1;
ⴱⴱⴱ ⫽ P ⬍ 0.0005 for LymphoStat-B versus the corresponding dose of human IgG1; # ⫽ P ⬍ 0.05 for
recombinant human BLyS versus buffer. hu ⫽ human; Ab ⫽ antibody.

of a 5-fold molar excess of LymphoStat-B, but not LymphoStat-B inhibition of the pharmacologic
control IgG, blocked BLyS binding. Taken together, effects of human BLyS in mice. Although human and
these findings demonstrate that LymphoStat-B blocks murine BLyS share significant amino acid homology
BLyS binding to cells via TACI, BCMA, and BR-3. (63% identity), LymphoStat-B does not neutralize the
LYMPHOSTAT-B, HUMAN MONOCLONAL ANTIBODY AGAINST BLyS 3261

Table 2. Effect of LymphoStat-B on tissue immunophenotype in cynomolgus monkeys*


MNCs analyzed,
study day,
LymphoStat-B dose CD3⫹ CD3⫹/CD4⫹ CD3⫹/CD8⫹ CD3⫺/CD14⫹ CD20⫹ CD20⫹/CD21⫹
Spleen MNCs
Day 29 (n ⫽ 6/group)
None (control) 25.9 ⫾ 9.3 8.8 ⫾ 3.9 8.2 ⫾ 4.3 1.04 ⫾ 0.6 51.5 ⫾ 13.0 10.7 ⫾ 5.3
5 mg/kg/dose 45.6 ⫾ 9.0† 16.9 ⫾ 5.4† 16.2 ⫾ 3.6 0.45 ⫾ 0.3 24.9 ⫾ 16.7† 6.2 ⫾ 5.4
15 mg/kg/dose 48.4 ⫾ 11.7† 15.0 ⫾ 5.5 15.5 ⫾ 6.9 0.65 ⫾ 0.4 30.5 ⫾ 15.0 6.0 ⫾ 4.6
50 mg/kg/dose 44.1 ⫾ 19.8 11.9 ⫾ 4.4 14.3 ⫾ 7.7 1.31 ⫾ 0.7 33.9 ⫾ 12.9 6.4 ⫾ 4.5
Day 57 (n ⫽ 4/group)
None (control) 33.6 ⫾ 10.1 15.2 ⫾ 4.0 13.6 ⫾ 3.1 2.21 ⫾ 1.1 39.3 ⫾ 12.6 8.2 ⫾ 0.9
5 mg/kg/dose 44.8 ⫾ 4.3 21.6 ⫾ 2.6 17.5 ⫾ 3.1 1.92 ⫾ 0.8 18.0 ⫾ 5.7† 3.1 ⫾ 0.7†
15 mg/kg/dose 41.9 ⫾ 10.6 18.3 ⫾ 7.2 18.5 ⫾ 4.0 1.72 ⫾ 1.1 22.5 ⫾ 7.7† 4.9 ⫾ 2.1†
50 mg/kg/dose 55.3 ⫾ 5.5† 23.8 ⫾ 3.3 24.7 ⫾ 2.6† 1.02 ⫾ 0.5 20.8 ⫾ 7.5† 3.5 ⫾ 1.4†

Lymph node MNCs


Day 29 (n ⫽ 6/group)
None (control) 67.0 ⫾ 6.7 46.6 ⫾ 6.3 18.2 ⫾ 5.6 1.18 ⫾ 0.6 28.7 ⫾ 9.2 5.2 ⫾ 1.9
5 mg/kg/dose 77.9 ⫾ 4.5† 56.0 ⫾ 5.6† 20.9 ⫾ 5.9 0.66 ⫾ 0.6 19.7 ⫾ 3.9 3.2 ⫾ 1.6
15 mg/kg/dose 82.3 ⫾ 6.0† 60.6 ⫾ 2.0† 20.7 ⫾ 5.8 0.43 ⫾ 0.1 16.0 ⫾ 4.5 3.4 ⫾ 1.6
50 mg/kg/dose 78.1 ⫾ 7.4† 55.5 ⫾ 4.9† 21.7 ⫾ 4.6 1.19 ⫾ 1.7 18.2 ⫾ 7.1 2.4 ⫾ 1.8
Day 57 (n ⫽ 4/group)
None (control) 70.9 ⫾ 7.9 47.9 ⫾ 8.0 22.3 ⫾ 3.6 1.66 ⫾ 0.4 26.7 ⫾ 5.9 10.8 ⫾ 3.8
5 mg/kg/dose 79.5 ⫾ 8.3 59.4 ⫾ 6.7 21.4 ⫾ 3.1 0.78 ⫾ 0.4 15.9 ⫾ 6.3† 5.0 ⫾ 2.7†
15 mg/kg/dose 84.7 ⫾ 2.3† 61.2 ⫾ 3.4† 25.1 ⫾ 7.7 1.72 ⫾ 0.7 11.5 ⫾ 3.0† 4.0 ⫾ 0.8†
50 mg/kg/dose 87.5 ⫾ 1.4† 59.4 ⫾ 5.3 30.0 ⫾ 6.0 0.69 ⫾ 0.7 11.2 ⫾ 1.9† 2.9 ⫾ 1.2†

* LymphoStat-B was administered at the indicated doses once every 7 days for 4 treatment cycles. Lymph node and spleen mononuclear cells
(MNCs) were analyzed by flow cytometry using the indicated markers. Values are the mean ⫾ SD percentage of spleen or lymph node MNCs.
† P ⬍ 0.05 versus control group, by Dunnett’s test (analysis of variance).

effects of murine BLyS (Human Genome Sciences: sults demonstrate that LymphoStat-B is an effective in
unpublished data) and is therefore unable to antagonize vivo antagonist of human BLyS bioactivity.
endogenous BLyS activity in either normal mice or Effects of administration of LymphoStat-B to
murine models of autoimmune disease. In order to cynomolgus monkeys. The in vivo consequences of BLyS
assess the in vivo efficacy of human-specific BLyS an- inhibition was evaluated in cynomolgus monkeys. Cyno-
tagonists, a mouse model was developed in which the molgus monkeys were considered a suitable animal
ability of LymphoStat-B to inhibit the pharmacologic model since cynomolgus BLyS is 96% identical to hu-
effects of exogenously administered human BLyS was man BLyS, and LymphoStat-B inhibits the in vitro
tested. This model allows the biologic actions of BLyS to activity of cynomolgus monkey BLyS with similar effi-
be measured as increases in spleen weight, increases in ciency as human BLyS (Human Genome Sciences: un-
serum IgA levels, and increases in the population of published data). Animals were injected with 5, 15, or 50
mature B cells in the spleen (B220⫹/ThB⫹ splenocytes; mg/kg of LymphoStat-B or vehicle once every 7 days for
the human equivalent of the markers are CD45R and 4 treatment cycles. Animals were necropsied on study
Ly6D, respectively) (1,35). day 29 or, following a 28-day treatment-free recovery
As shown in Figure 7, subcutaneous administra- period, on day 57.
tion of 0.3 mg/kg of human BLyS for 4 consecutive days Consistent with the phenotype of BLyS-deficient
resulted in increases in spleen weights, in the represen- mice (18,19), a decrease in B cell representation was
tation of CD45R⫹ (B220⫹)/ThB⫹ splenocytes, and in observed in spleen and mesenteric lymph nodes from
the total serum IgA concentrations. Coadministration of LymphoStat-B–treated monkeys on both day 29 and day
LymphoStat-B intravenously resulted in a dose-dependent 57, as monitored by flow cytometry (Table 2). As might
inhibition of these human BLyS-induced effects, with be expected, there was a reciprocal increase in the
complete inhibition observed between 1.5 and 5.0 mg/kg percentage of tissue T cells, but there was no corre-
of monoclonal antibody. No inhibition of BLyS-induced sponding increase in circulating T cells, nor was there a
effects was achieved by administration of the Ig isotype consistent T cell immunophenotype that was increased
control monoclonal antibody (human IgG1). These re- (i.e., CD8⫹ versus CD4⫹). Therefore, it is likely that
3262 BAKER ET AL

the observed increase in the relative percentage of T syndrome (25), consistent with clinical observations of
cells simply reflects the B cell depletion in these tissues. the association of Sjögren’s syndrome in subsets of
Consistent with this, histologic analysis of these tissues patients with SLE (39). Taken together, these observa-
demonstrated a mild lymphoid depletion primarily in B tions suggest that elevated levels of BLyS may contribute
cell–dependent areas (data not shown). No significant to B cell hyperactivity and autoantibody production in
changes were observed in peripheral blood mononuclear multiple autoimmune diseases by enhancing the survival
cells detected by flow cytometry or in serum Ig concen- of B cells. Specific targeting of BLyS may therefore
trations (data not shown). These results demonstrate provide a novel treatment for these diseases. Toward
that LymphoStat-B is able to inhibit BLyS in vivo and that end, we have generated a human antibody specific
that this inhibition results in depletion of B cell popula- for human BLyS that can neutralize the biologic func-
tions after a relatively short course of treatment. tions of BLyS in vitro and in vivo.
LymphoStat-B was generated from a naive hu-
DISCUSSION man Ig library using phage display technology. More
than 1,200 antibodies with distinct amino acid sequences
BLyS plays a critical role in the normal regulation were isolated that specifically bound soluble human
of B cell development and immune response. This was BLyS. Optimization of selected main candidates allowed
demonstrated by the severely depleted B cell phenotype further improvements in activity, leading to the selection
observed in BLyS-deficient animals (18,19). As is the of the clinical development candidate, LymphoStat-B.
case for other cytokines, BLyS levels must be carefully Interactions between BLyS and its receptors are
regulated in order to maintain “immune homeostasis” complex. At least 3 distinct receptors exist for BLyS. The
(37). Consistent with this notion are findings that over- major receptor for mediating BLyS-dependent B cell
production of BLyS is associated with increased immu- development appears to be the BLyS-specific receptor
noglobulin production and autoimmunity. Transgenic BR-3. This is based on data showing that the A/WySnJ
mice overexpressing either murine or human BLyS show mouse strain, which has a defect in BR-3, mirrors the
increased numbers of mature B cells in the spleen and severe deficits in peripheral mature B cell populations
lymph nodes, elevated levels of total immunoglobulins, seen in BLyS-deficient mice (9,10,18–20). Although
rheumatoid factor, anti–double-stranded DNA antibod- BLyS binds to 2 additional receptors, BCMA and TACI,
ies, and circulating immune complexes, all of which are analyses of mice deficient in these receptors show no B
manifestations associated with autoimmune disease. The cell deficits in the BCMA knockout mice (14) and
mice eventually develop proteinuria and glomerulone- increased levels of B cells in TACI-deficient mice (15),
phritis, symptoms that are specifically associated with suggesting a redundant role for BCMA and an inhibitory
SLE (21,22). Elevated levels of BLyS have also been role for TACI in the regulation of B cell maturation
detected in the (NZB ⫻ NZW)F1 and MRL-lpr/lpr (17). Importantly, we show by 2 independent methods
mouse models of lupus. BLyS levels in these animals that LymphoStat-B is able to neutralize BLyS interac-
increase with age, in parallel with increased autoanti- tions with all 3 BLyS receptors.
body production, proteinuria, and other disease manifes- BLyS, like many TNF family members, exists in
tations, again suggesting an important role for BLyS in the both soluble and membrane-bound forms. Most avail-
development of autoimmune disease in animals (6). able data support the action of BLyS as a soluble
Evidence that BLyS is associated with human cytokine (1), and elevated levels of soluble BLyS are
autoimmune disease comes from several studies demon- correlated with autoimmune disease (23,24). Any spe-
strating elevated levels of circulating BLyS in serum cific activity of membrane-bound BLyS remains to be
samples from patients with SLE and RA (23,24). In RA determined. In fact, the exact nature of membrane-
patients, elevated levels of BLyS were also found in bound BLyS is not fully understood, but it has been
synovial fluid and were consistently greater than the shown that a panel of antibodies can differentially
serum levels in the same patient, possibly suggesting that recognize membrane-bound BLyS (7). The BLyS anti-
there is local production of BLyS at sites of inflamma- bodies described in the present study were selected
tion (38). Increased BLyS levels have also been found in specifically for their ability to recognize and inhibit the
sera from patients with Sjögren’s syndrome, another effects of soluble human BLyS. LymphoStat-B does not
autoimmune disease characterized by B cell hyperreac- recognize membrane-bound BLyS and does not bind to
tivity and autoantibody production (25,26). Mice trans- the surface of human cells that bear membrane-bound
genic for BLyS also develop symptoms of Sjögren’s BLyS. Thus, the specific activity of soluble BLyS is
LYMPHOSTAT-B, HUMAN MONOCLONAL ANTIBODY AGAINST BLyS 3263

targeted, and any nonspecific effects that might be comitant effects on serum immunoglobulin levels (42), it
associated with antibody binding to BLyS-producing is perhaps not surprising that changes in serum immu-
cells should be prevented. noglobulins were not observed in the study reported
BLyS has also been shown to exist in a complex here. Although the effects of LymphoStat-B administra-
with APRIL to form heterotrimers. Although the bio- tion over a short period of time might not be expected to
logic significance of these heterotrimers is not clearly fully reflect the phenotypes of BLyS knockout mice, one
defined, they have in vitro biologic activity and interact key similarity with the BLyS knockout mice, namely a
with BLyS receptors (40). While LymphoStat-B was reduction of tissue B cell populations, was observed
raised against soluble “homotrimeric” BLyS, interac- following drug administration.
tions with heterotrimers have been shown in in vitro LymphoStat-B possesses many properties that
assays (Smith R, Roschke V: unpublished observations), make it ideally suited for use as a therapeutic agent.
although the affinity of these interactions was 7–8-fold First, it binds with high affinity to soluble human BLyS
lower than that of the heterotrimer recognizing antibod- and inhibits its activities both in vitro and in vivo.
ies reported previously. The significance of this inhibi- Second, it does not recognize other TNF family mem-
tion awaits a clearer delineation of the in vivo role of bers, including its closest homolog human APRIL, and is
BLyS/APRIL heterotrimers in B cell regulation. therefore specific for the factor implicated in the patho-
In an in vitro assay, LymphoStat-B was found to genesis of several autoimmune diseases. As an antibody,
inhibit human BLyS-induced stimulation of B cells from an additional advantage of LymphoStat-B is its long
both murine splenocytes and human tonsillar B cells terminal half-life. In mice and monkeys, the half-life of
(data not shown). In vivo, LymphoStat-B neutralizes the LymphoStat-B is 2.5 days and 11–14 days, respectively
observable effects of exogenously administered human (Riccobene T: unpublished observations).
BLyS in mice. These effects include stimulation of B cell Overproduction of BLyS has been associated
proliferation, as evidenced by increases in spleen weight, with SLE, RA, and Sjögren’s syndrome. The etiologies
representation of mature marginal zone B cells, as of these diseases are unknown, but they share the
demonstrated by increases in B220⫹/ThB⫹ B cells, and common features of B cell hyperactivity and autoanti-
immunoglobulin secretion, as evidenced by increases in body production, which are strongly implicated in the
IgA levels. LymphoStat-B at dosages of 1–5 mg/kg pathogenesis of the disease process. Support for the
completely prevented these BLyS-induced activities. In ability of an antagonist of BLyS to affect these diseases
this model system, LymphoStat-B has similar efficiencies comes from several animal studies with soluble BLyS
when administered via the intravenous or the subcuta- receptors. Administration of TACI-Fc or BCMA-Fc has
neous route (Hilbert D: unpublished observations). It been shown to inhibit antigen-specific antibody re-
should also be noted that the dosing schedule of 4 daily sponses and decrease B cell numbers and germinal
injections of BLyS used in the present study has only centers (43,44). TACI-Fc and BR-3-Fc have also been
modest effects on increasing IgM levels and little, if any, shown to inhibit the development of SLE disease man-
effects on IgG levels (35). Nevertheless, LymphoStat-B ifestations in the (NZB ⫻ NZW)F1 mouse (6,28).
can reduce BLyS-induced changes in IgM (data not TACI-Fc has also prevented disease onset in the
shown). collagen-induced arthritis model in mice (27).
Administration of LymphoStat-B to normal cyno- The critical role BLyS plays in the regulation of B
molgus monkeys resulted in tissue B cell depletion that cell homeostasis suggests that deregulation of BLyS may
was sustained for up to 4 weeks after the treatment underlie these as well as additional B cell–mediated
period. Although no significant depletion of circulating diseases. LymphoStat-B is being developed as a novel
B cells was observed at these time points, it has been treatment for autoimmune diseases and is currently
suggested that the spleen and lymphoid tissue act as being evaluated in a phase I clinical trial in SLE patients.
reservoirs for the supply of B cells into the bloodstream Studies are also under way to identify additional diseases
(41), and effects on circulating B cells may not be in which BLyS might play a role and LymphoStat-B may
observed until reserves of B cells in lymphoid tissues are have therapeutic potential.
adequately suppressed. No significant changes were
observed in serum immunoglobulin levels following
LymphoStat-B administration. However, since the B ACKNOWLEDGMENTS
cell–depleting drug rituximab results in severe (⬎90%) The authors wish to thank members of the DNA
and prolonged B cell depletion, with only minor con- sequencing facility at Human Genome Sciences for antibody
3264 BAKER ET AL

sequencing, Steve Barash for bioinformatics analysis, Steve normal development of B cells through a BCMA-independent
Ullrich for antibody purification, and Jeff Carrell, Ernest pathway. Science 2001;293:2111–14.
Boyd, and Dana Bresette for flow cytometry. 19. Gross JA, Dillon SR, Mudri S, Johnston J, Littau A, Roque R, et
al. TACI-Ig neutralizes molecules critical for B cell development
and autoimmune disease: impaired B cell maturation in mice
REFERENCES lacking BLyS. Immunity 2001;15:289–302.
20. Harless SM, Lentz VM, Sah AP, Hsu BL, Clise-Dwyer K, Hilbert
1. Moore PA, Belvedere O, Orr A, Pieri K, LaFleur DW, Feng P, et DM, et al. Competition for BLyS-mediated signaling through
al. BLyS: member of the tumor necrosis factor family and B Bcmd/BR3 regulates peripheral B lymphocyte numbers. Curr Biol
lymphocyte stimulator. Science 1999;285:260–3. 2001;11:1986–9.
2. Schneider P, MacKay F, Steiner V, Hofmann K, Bodmer JL, 21. Mackay F, Woodcock SA, Lawton P, Ambrose C, Baetscher M,
Holler N, et al. BAFF, a novel ligand of the tumor necrosis factor Schneider P, et al. Mice transgenic for BAFF develop lymphocytic
family, stimulates B cell growth. J Exp Med 1999;189:1747–56. disorders along with autoimmune manifestations. J Exp Med
3. Shu HB, Hu WH, Johnson H. TALL-1 is a novel member of the 1999;190:1697–710.
TNF family that is down-regulated by mitogens. J Leukoc Biol 22. Khare SD, Sarosi I, Xia XZ, McCabe S, Miner K, Solovyev I, et al.
1999;65:680–3. Severe B cell hyperplasia and autoimmune disease in TALL-1
4. Mukhopadhyay A, Ni J, Zhai Y, Yu G-L, Aggarwal BB. Identifi- transgenic mice. Proc Natl Acad Sci U S A 2000;97:3370–5.
cation and characterization of a novel cytokine, THANK, a TNF 23. Zhang J, Roschke V, Baker KP, Wang Z, Alarcón GS, Fessler BJ,
homologue that activates apoptosis, nuclear factor-␬B, and c-Jun et al. A role for B lymphocyte stimulator in systemic lupus
NH2-terminal kinase. J Biol Chem 1999;274:15978–81. erythematosus. J Immunol 2001;166:6–10.
5. Tribouley C, Wallroth M, Chan V, Paliard X, Fang E, Lamson G, 24. Cheema GS, Roschke V, Hilbert DM, Stohl W. Elevated serum B
et al. Characterization of a new member of the TNF family lymphocyte stimulator levels in patients with systemic immune-
expressed on antigen presenting cells. Biol Chem 1999;380:1443–7. based rheumatic diseases. Arthritis Rheum 2001;44:1313–9.
6. Gross JA, Johnston J, Mudri S, Enselman R, Dillon SR, Madden 25. Groom J, Kalled SL, Cutler AH, Olson C, Woodcock SA, Schnei-
K, et al. TACI and BCMA are receptors for a TNF homologue der P, et al. Association of BAFF/BLyS overexpression and altered
implicated in B-cell autoimmune disease. Nature 2000;404:995–9. B cell differentiation with Sjögren’s syndrome. J Clin Invest
7. Nardelli B, Belvedere O, Roschke V, Moore PA, Olsen HO, 2002;109:59–68.
Migone TS, et al. Synthesis and release of B lymphocyte stimulator 26. Mariette X, Roux S, Zhang J, Bengoufa D, Lavie F, Zhou T, et al.
from myeloid cells. Blood 2001;97:198–204. The level of BLyS (BAFF) correlates with the titre of autoanti-
8. Do RK, Hatada E, Lee H, Tourigny MR, Hilbert D, Chen-Kiang bodies in human Sjögren’s syndrome. Ann Rheum Dis 2003;62:
S. Attenuation of apoptosis underlies B lymphocyte stimulator 168–71.
enhancement of humoral immune response. J Exp Med 2000;192: 27. Wang H, Marsters SA, Baker T, Chan B, Lee WP, Fu L, et al.
953–64. TACI-ligand interactions are required for T cell activation and
9. Thompson JS, Bixler SA, Qian F, Vora K, Scott ML, Cachero TG, collagen-induced arthritis in mice. Nat Immunol 2001;2:632–7.
et al. BAFF-R, a newly identified TNF receptor that specifically 28. Kayagaki N, Yan M, Seshasayee D, Wang H, Lee W, French DM,
interacts with BAFF. Science 2001;293:2108–11. et al. BAFF/BLyS receptor 3 binds the B cell survival factor BAFF
10. Yan M, Brady JR, Chan B, Lee WP, Hsu B, Harless S, et al. ligand through a discrete surface loop and promotes processing of
Identification of a novel receptor for B lymphocyte stimulator that NF-␬B2. Immunity 2002;10:515–24.
is mutated in a mouse strain with severe B cell deficiency. Curr 29. Vaughan TJ, Williams AJ, Pritchard K, Osbourn JK, Pope AR,
Biol 2001;11:1547–52. Earnshaw JC, et al. Human antibodies with sub-nanomolar affin-
11. Marsters SA, Yan M, Pitti RM, Haas PE, Dixit VM, Ashkenazi A. ities isolated from a large non-immunized phage display library.
Interaction of the TNF homologues BLyS and APRIL with the Nat Biotechnol 1996;14:309–14.
TNF receptor homologues BCMA and TACI. Curr Biol 2000;10: 30. Hawkins RE, Russell SJ, Winter G. Selection of phage antibodies
785–8. by binding affinity: mimicking affinity maturation. J Mol Biol
12. Wu Y, Bressette D, Carrell JA, Kaufman T, Feng P, Taylor K, et 1992;266:889–96.
al. Tumor necrosis factor receptor superfamily member TACI is a 31. McCafferty J, Fitzgerald KJ, Earnshaw J, Chiswell DJ, Link J,
high affinity receptor for TNF family members APRIL and BLyS. Smith R, et al. Selection and rapid purification of murine antibody
J Biol Chem 2000;275:35478–85. fragments that bind a transition-state analog by phage display.
13. Yu G, Boone T, Delaney J, Hawkins N, Kelley M, Ramakrishnan Appl Biochem Biotechnol 1994;47:157–73.
M, et al. APRIL and TALL-1 and receptors BCMA and TACI: 32. Gibson T. Studies on the Epstein-Barr virus genome [thesis].
system for regulating humoral immunity. Nat Immunol 2000;1: Cambridge: University of Cambridge; 1984.
252–6. 33. Griffiths AD, Malmqvist M, Marks JD, Bye JM, Embleton MJ,
14. Xu S, Lam KP. B-cell maturation protein, which binds the tumor McCafferty J, et al. Human anti-self antibodies with high specific-
necrosis factor family members BAFF and APRIL, is dispensable ity from phage display libraries. EMBO J 1993;12:725–34.
for humoral immune responses. Mol Cell Biol 2001;21:4067–74. 34. Persic L, Roberts A, Wilton J, Cattaneo A, Bradbury A, Hoogen-
15. Von Bulow G-U, van Deursen JM, Bram RJ. Regulation of the boom HR. An integrated vector system for the eukaryotic expres-
T-independent humoral response by TACI. Immunity 2001;14: sion of antibodies or their fragments after selection from phage
573–82. display libraries. Gene 1997;187:9–18.
16. Yan M, Wang H, Chan B, Roose-Girma M, Erickson S, Baker T, 35. Parry TJ, Riccobene TA, Strawn SJ, Williams R, Daoud R, Carrell
et al. Activation and accumulation of B cells in TACI-deficient J, et al. Pharmacokinetics and immunological effects of exog-
mice. Nat Immunol 2001;2:638–43. enously administered recombinant human B lymphocyte stimula-
17. Seshasayee D, Valdez P, Yan M, Dixit VM, Tumas D, Grewal IS. tor (BLyS) in mice. J Pharmacol Exp Ther 2001;296:396–404.
Loss of TACI causes fatal lymphoproliferation and autoimmunity, 36. Kijek TM, Rossi CA, Moss D, Parker RW, Henchal EA. Rapid
establishing TACI as an inhibitory BLyS receptor. Immunity and sensitive immunomagnetic-electrochemiluminescent detec-
2003;18:279–88. tion of staphylococcal enterotoxin B. J Immunol Methods 2000;
18. Schiemann B, Gommerman JL, Vora K, Cachero TG, Shulga- 236:9–17.
Morskaya S, Dobles M, et al. An essential role for BAFF in the 37. Dy M, Vazquez A, Bertoglio J, Theze J. General aspects of
LYMPHOSTAT-B, HUMAN MONOCLONAL ANTIBODY AGAINST BLyS 3265

cytokine properties and functions. In: Theze J, editor. The cyto- 41. Kreuzfelder E, Obertacke U, Erhard J, Funk R, Steinen R,
kine network and immune functions. Oxford: Oxford University Scheiermann N, et al. Alterations of the immune system following
Press; 1999. p. 1–13. splenectomy in childhood. J Trauma 1991;31:358–64.
38. Tan S-M, Xu D, Roschke V, Perry JW, Arkfeld DG, Ehresmann 42. Maloney DG, Grillo-Lopez AJ, White CA, Bodkin D, Schilder RJ,
GR, et al. Local production of B lymphocyte stimulator protein Neidhart JA, et al. IDEC-C2B8 (rituximab) anti-CD20 monoclo-
and APRIL in arthritic joints of patients with inflammatory nal antibody therapy in patients with relapsed low-grade non-
arthritis. Arthritis Rheum 2003;48:982–92. Hodgkin’s lymphoma. Blood 1997;90:2188–95.
39. Jonsson R, Haga H-J, Gordon TP. Sjögren’s syndrome. In: Koop- 43. Schneider P, Takatsuka H, Wilson A, Mackay F, Tardivel A, Lens
man WJ, editor. Arthritis and allied conditions: a textbook of S, et al. Maturation of marginal zone and follicular B cells requires
rheumatology. 14th ed. Philadelphia: Lippincott Williams & B cell activating factor of the tumor necrosis factor family and is
Wilkins; 2001. p. 1736–59. independent of B cell maturation antigen. J Exp Med 2001;194:
40. Roschke V, Sosnovtseva S, Ward CD, Hong JS, Smith R, Albert V, 1691–7.
et al. BLyS and APRIL form biologically active heterotrimers that 44. Yan M, Marsters SA, Grewal IS, Wang H, Ashkenazi A, Dixit VM.
are expressed in patients with systemic immune-based rheumatic Identification of a receptor for BLyS demonstrates a crucial role in
diseases. J Immunol 2002;169:4314–21. humoral immunity. Nat Immunol 2000;1:37–41.

You might also like