Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

IMMUNOBIOLOGY

Anti-BR3 antibodies: a new class of B-cell immunotherapy combining cellular


depletion and survival blockade
Wei Yu Lin,1 Qian Gong,1 Dhaya Seshasayee,1 Zhonghua Lin,1 Qinglin Ou,1 Shiming Ye,1 Eric Suto,1 Jean Shu,1
Wyne Pun Lee,1 Ching-Wei V. Lee,2 Germaine Fuh,2 Maya Leabman,3 Suhasini Iyer,3 Kathy Howell,3 Thomas Gelzleichter,3
Joseph Beyer,3 Dimitry Danilenko,4 Sherry Yeh,5 Laura E. DeForge,5 Allen Ebens,6 Jeffrey S. Thompson,7
Christine Ambrose,7 Mercedesz Balazs,1 Melissa A. Starovasnik,2 and Flavius Martin1
Departments of 1Immunology, 2Protein Engineering, 3Development Sciences, 4Pathology, 5Assay and Automation Technology, and 6Translational Oncology,
Genentech, South San Francisco, CA; and 7Department of Gene Discovery, Biogen IDEC, Cambridge Center, Cambridge, MA

Removal of pathogenic B lymphocytes by being applied to develop novel antibodies with BR3-Fc BAFF blockade or with anti-
depletion of monoclonal antibodies with additional functional attributes. We CD20 treatment. Comparative analysis of
(mAbs) or deprivation of B-cell survival describe the generation of a novel class BR3-Fc and anti-BR3 mAb reveals a lower
factors has demonstrated clinical benefit of B-cell-directed therapy (anti-BR3 mAbs) B-cell dependence for BAFF-mediated
in both oncologic and immunologic dis- that combines the depleting capacity of a survival in nonhuman primates than in
eases. Partial clinical responses and therapeutic mAb and blockade of B-cell- mice. This novel class of B-cell-targeted
emerging data demonstrating incomplete activating factor (BAFF)–BR3 B-cell sur- therapies shows species characteristics
B-cell depletion after immunotherapy fu- vival. In mice, treatment with antagonistic in mice and primates that will guide trans-
els the need for improved therapeutic anti-BR3 antibodies results in quantita- lation to treatment of human disease.
modalities. Lessons from the first genera- tively greater reduction in some B-cell (Blood. 2007;110:3959-3967)
tion of therapeutics directed against B- subsets and qualitatively different effects
cell-specific antigens (CD20, CD22) are on bone marrow plasma cells compared © 2007 by The American Society of Hematology

Introduction
B-cell immunotherapy has shown clinical benefit in the treatment (ADCC), complement-mediated cytotoxicity (CDC), and apoptosis
of human B-cell malignancies and autoimmune diseases.1-4 Since have been demonstrated as the main mechanisms of action in a
approval of a depleting anti-CD20 monoclonal antibody (mAb) variety of systems including mouse xenotumors and normal mouse
(rituximab [Genentech, South San Francisco, CA; Biogen-IDEC, and nonhuman primate (NHP) B-cell subsets.11-15 Two major
Cambridge, MA; Roche, Basel, Switzerland)] in 1997 for the determinants affecting normal mouse B-cell depletion have been
treatment of non-Hodgkin lymphoma (NHL), almost 1 million identified.13,16 First, the kinetics of B-cell recirculation determines
patients have been treated with rituximab as a first or second line the speed and magnitude of anti-CD20 mAb-mediated B-cell
therapy, either alone or in combination with chemotherapy. In depletion. Cells with higher recirculatory kinetics from blood,
addition, rituximab maintenance therapy significantly prolongs lymph nodes, and spleen follicular areas are depleted faster and
tumor remission and patient survival in patients with indolent more completely than cells with lower recirculatory kinetics (eg,
B-cell NHL or chronic lymphocytic leukemia (CLL).5,6 More peritoneal cavity [PEC], marginal zone [MZ], germinal centers
recently, rituximab has demonstrated clinical benefit in a variety of [GC]). Second, the local microenvironment influences the extent of
autoimmune diseases including rheumatoid arthritis, pemphigus B-cell depletion. Marginal zone, Peyer patches (PP), germinal
vulgaris, immune thrombocytopenia and autoimmune hemolytic center, memory, and peritoneal cavity B cells exhibit greater
anemia.4,7,8 As a result, understanding the contribution of B resistance to depletion in mice and nonhuman primates. Reduced
lymphocytes to human autoimmune diseases received revived recruitment of effector mechanisms in the peritoneal cavity as well
interest, and several mechanisms have been postulated to partici- as intrinsic B1 B cells properties appear to cause the slower kinetics
pate in disease pathogenesis, including autoantibody production, and B-cell reduction after anti-CD20 mAb treatment.16 Differences
B-cell antigen presentation, cytokine generation, and lymphorgano- across mouse strains and epitopes recognized by anti-CD20
genesis.2,3,9,10 Inhibition of different combinations of these mecha- antibodies used for depletion might explain the differential effects
nisms is probably responsible for clinical benefit. seen on mouse splenic marginal zone B cells.13,16 Small but
The success of anti-CD20 B-cell immunotherapy spearheaded a consistent numbers of residual B cells can be detected in most
large number of preclinical and clinical efforts to understand in lymphoid organs in mice and primates treated with anti-CD20
vivo mechanisms of drug activity. Direct elimination of malignant mAbs. One possibility of achieving a more complete B-cell
B cells through antigen-dependent cell-mediated cytotoxicity reduction would be to block B-cell survival signals in addition to

Submitted April 30, 2007; accepted July 31, 2007. Prepublished online as The online version of this article contains a data supplement.
Blood First Edition Paper, August 8, 2007; DOI 10.1182/blood-2007-
The publication costs of this article were defrayed in part by page charge
04-088088.
payment. Therefore, and solely to indicate this fact, this article is hereby
marked ‘‘advertisement’’ in accordance with 18 USC section 1734.
An Inside Blood analysis of this article appears at the front of this issue. © 2007 by The American Society of Hematology

BLOOD, 1 DECEMBER 2007 䡠 VOLUME 110, NUMBER 12 3959


3960 LIN et al BLOOD, 1 DECEMBER 2007 䡠 VOLUME 110, NUMBER 12

administration of a B-cell-depleting reagent. B-cell-activating isotype control in all mouse studies. Final endotoxin levels of antibodies for
factor (BAFF) (also known as BLyS) and a proliferation-inducing all in vivo studies were less than 0.2 EU/mg. To be able to compare among
ligand (APRIL), 2 related tumor necrosis factor (TNF) family molecules, mice were dosed in all experiments to ensure maximal B-cell
members, and their receptors—BLyS receptor 3 (BR3; also known reduction based on in vivo titrations of anti-BR3, BR3-Fc and anti-CD20
as BAFFR), transmembrane activator and calcium modulator and reagents. Dose regimes were determined to achieve similar trough concen-
trations for anti-BR3 and BR3-Fc in all experiments. Anti-BR3 at a 500-␮g
cyclophilin ligand interactor (TACI), and B-cell maturation antigen
single dose achieves blood drug levels at day 15 similar to 200-␮g weekly
(BCMA)—constitute a well characterized system involved in
multidose trough concentrations. BR3-Fc at 150 ␮g, 3 times per week,
B-cell development and survival.17-21 Of these, BAFF/BR3 is the
achieves a 72-hour trough in a similar range. Target trough concentrations
main axis transducing B-cell survival signals, and interference with were determined to achieve maximal B-cell reduction in splenic B-cell
this pathway using blocking reagents (anti-BAFF antibodies or subsets based on a series of single-dose pharmacokinetic/pharmacodynamic
BR3-, TACI-, or BCMA-Fc fusion proteins) results in B-cell experiments. For example, at day 4, anti-BR3 blood concentrations reached
reductions in mice, nonhuman primates, and humans.22,23 Recent after a 20-␮g dose did not achieve maximal B-cell depletion, whereas those
clinical trials with an anti-BAFF antibody have shown clinical reached after a 200-␮g dose did (Figure S1, available on the Blood website;
benefit in rheumatoid arthritis (RA) and positive trends in retrospec- see the Supplemental Materials link at the top of the online article). The
tive subset analysis in patients with systemic lupus erythematosus half-life is 5 days for anti-CD20 and anti-BR3 antibodies and
(SLE).24,25 In mice, BAFF blockade with BR3-Fc appears to act 3 days for mouse BR3-Fc. Mouse anti-human BR3 (clone 9.1; Biogen-
synergistically with anti-CD20, resulting in more complete B-cell IDEC49) was humanized on a human IgG1 framework and used in NHP
depletion at doses suboptimal as monotherapy.13 It is noteworthy studies. All in vitro properties (eg, BR3 binding, BAFF antagonism, ADCC)
that serum BAFF levels increase when B cells are lacking (ie, were similar between the parent and humanized antibody.
rag-deficient mice) or after depletion with rituximab in patients
with RA, SLE, or Sjögren syndrome, probably because of a Animals
decrease in receptor-mediated clearance and possibly other mecha-
nisms.26-30 Thus, increased serum BAFF could provide additional Eight- to 12-week-old BALB/c mice (The Jackson Laboratory, Bar Harbor,
Maine) and C57BL/6 mice (Charles River Laboratories, Wilmington, MA)
survival signals for cells that were not removed during the
were used for depletion studies. hCD20tgBAC FvB and C57BL/6 mice
depletion phase and for emerging new B cells during the recovery
were described previously.13 NZB/W F1 female mice (The Jackson
phases of B-cell immunotherapy.
Laboratory), an SLE-prone model, were ⬃7 to 8 months old and weighed
Strategies targeted at enhancing intrinsic properties of anti- 35 to 50 g when they were enrolled in therapeutic studies. Urine was tested
CD20 antibodies (eg, increased ADCC, CDC, and affinity to for proteinuria by semiquantitatively dipsticks (Multistick; Thermo Fisher
antigen) represent one modality by which to achieve greater Scientific, Waltham, MA) every 4 weeks. The baseline protein levels at the
clinical benefit compared with existing B-cell immunotherapy.31-35 start of the studies were between 30 and 300 mg/dL. All experimental
Other efforts are driven toward testing antibodies directed against animal studies were conducted according to protocols that were reviewed
other B-cell surface molecules (CD19, CD22).36-39 Finally, conju- and approved by the Institutional Animal Care and Use Committees
gates of radioactive isotypes (ibritumomab tiuxetan [Zevalin], (IACUC) of Genentech Lab Animal Research (LAR).
tositumomab [Bexxar]) and toxins (calicheamicin-antiCD22, anti- NHP cynomolgus monkey studies were conducted at contract research
CD79 antibody-drug conjugates) represent a third strategy to organizations (CROs) according to their standard operating procedures and
enhance B-cell targeting.40-42 In addition to intrinsic molecule in compliance with applicable regulations concerning the use of laboratory
improvements, combinations of B-cell-depleting agents with animals. Naive monkeys, 2 to 5 years old, were acclimated to the study
molecules enhancing other arms of the immune system in- rooms for 28 days before the initiation of the study.
cluding effector mechanisms for B-cell depletion (eg, interferon-␣,
granulocyte–colony-stimulating factor, granulocyte macrophage– Tissue collection
colony-stimulating factor, interleukin 2) are being attempted in
Whole spleen, lymph nodes, Peyer patches, and kidneys were collected for
B-cell malignancies with variable success.43-46
analysis. Single-cell suspensions were prepared by grinding organs between
Based on the demonstrated synergy between B-cell depletion
frosted glass slides. The total cell counts were measured using fluorescence
and blockade of B-cell survival through BR3, we developed beads. Cells were filtered through a 70-␮m nylon cell strainer and used for
anti-BR3 antibodies that combine, in one molecule, the abilities to flow cytometry analysis and spot enzyme-linked immunosorbent assays.
deplete B cells and to block BAFF-mediated B-cell survival. These Blood samples were collected from the retro-orbital sinus (100 ␮L) at
antibodies combine 2 mechanisms of action and show increased in trough and by cardiac puncture (while the animals were anesthetized with
vivo B-cell reduction in some subsets compared with anti-CD20 3% isoflurane anesthesia, 600⬃800 ␮L) before the mice were euthanized.
mAb-mediated B-cell depletion or BAFF-dependent survival block-
ade on their own. As a result, therapy with this novel type of
Flow cytometry and antibodies
antibody has the potential to translate into increased clinical benefit
in diseases with B-cell-mediated pathogenic mechanisms. Spleen, lymph nodes, Peyer patches, peritoneal cavity lavage, and periph-
eral blood cells were analyzed for B and T cell subsets using conjugated
antibodies as fluorescence-activated cell sorting (FACS) reagents: anti-
mouse CD3, CD4, CD8, CD5, CD23, CD21, CD69, B220, CD38, CD138,
Materials and methods CD44, CD62L, and anti-human CD19, CD5, and cynomolgus monkey
cross-reactive CD20, CD40, CD21, and CD27, all from Pharmingen (San
Anti-BR3 antibodies and in vivo dosing
Diego, CA) and BD Biosciences (San Jose, CA). Polyclonal goat anti-
mCB2 (mIgG2a) and mCB2 DANA, phage-displayed synthetic antibody, mouse IgM, IgG1, IgG2a, and IgA were purchased from Southern
were generated as described previously.47 PIH11 (anti-mouse BR3, nonblock- Biotechnology Associates (Birmingham, AL). FACS analyses were con-
ing antibody; Biogen-IDEC, Cambridge, MA), mBR3-Fc, and mouse ducted on a FACSCalibur (BD Biosciences). B-cell subsets were identified
anti-human CD20 (mouse IgG2a version of anti-human CD20 mAb 2H7) as described in each figure legend. Statistical analyses relative to the control
were described previously.13,48 Anti-ragweed mouse IgG2a was used as the group were performed using the Dunnett test.
BLOOD, 1 DECEMBER 2007 䡠 VOLUME 110, NUMBER 12 IN VIVO B CELL EFFECTS OF ANTI-BR3 ANTIBODIES 3961

Figure 1. Anti-BR3 mAb reduces mouse B-cell subsets more efficiently than BR3-Fc. BALB/c mice were treated with anti-BR3 (500 ␮g), BR3-Fc (150 ␮g ⫻ 3/week) or
isotype control (500 ␮g); 15 days later, tissues were analyzed by flow cytometry (A,C) and immunofluorescence (B). Percentages of cells in B-cell subsets are displayed near
each gate as follows: Spleen: follicular (FO) B cells (CD23⫹CD21⫹), marginal zone (MZ) B cells (CD23⫺CD21hi), and plasma cells (PC, CD138⫹B220lo); Lymph nodes: B cells
(CD23⫹CD21⫹), blood (BL) B cells (CD23⫹CD21⫹), Peyer patches (PP), follicular B cells (B220⫹CD38⫹), and germinal center (GC) B cells (B220⫹CD38lo); peritoneal cavity
(PEC): B2 cells (B220⫹CD23⫹) and B1 cells (B220⫹CD23⫺) (A,C). B cells (blue) and T cells (green) are stained in spleen sections after treatment with control, BR3-Fc or
anti-BR3 mAb. Marginal zone and follicular B-cell areas are separated by the layer of metallophilic macrophages (MM [red]) (B). See “Immunofluorescence and
immunohistochemistry staining” for image acquisition details. Absolute B-cell numbers or percentage of lymphocyte gate belonging to a defined subset are shown as mean plus
or minus standard error. Statistical significance compared with the control treated group is indicated with asterisk (C). When 2 treatment groups are significantly different from
each other, the P value is displayed. These data are representative of more than 10 independent experiments done with several maximally depleting doses of anti-BR3, using
at least 5 mice/group.

BrdU labeling & staining plished using mouse IgG2a anti-human CD20 (clone L26; Dako,
Carpinteria, CA) or an irrelevant IgG2a antibody (clone C1.18.4; BD
One week before take down, each mouse was received 2 mg of bromode-
Pharmingen) and detection system using ABC-Peroxidase Elite with
oxyuridine (BrdU; Sigma, St. Louis, MO) in phosphate-buffered saline
Vector Blue (Vector Laboratories). Immunohistochemistry for T cells
(PBS) via intraperitoneal (i.p.) injection every other day for a week.
was accomplished using mouse IgG1 anti-CD3 (clone SP34–2; BD
BrdU staining was performed using a BrdU flow kit (BD Biosciences).
Pharmingen) or an irrelevant IgG1 antibody (clone MOPC-21; BD
First, B lineage surface antigens B220 and CD138 were stained, followed
by fixation and permeabilization of cells using BD Cytofix/Cytoperm Pharmingen) and detection system using ABD-Peroxidase Elite with
buffer. The cells were then treated with DNase to expose BrdU epitopes. diaminobenzidine (Vector Laboratories).
Cycling S phase cells were detected by staining cells with fluorochrome
conjugated anti-BrdU and anti-Ig isotype antibodies and then analyzing by Anti-DNA spot enzyme-linked immunosorbent assay
flow cytometry.
Sheared calf thymus DNA (Invitrogen, Carlsbad, CA) was coated
Immunofluorescence and immunohistochemistry staining overnight onto 96-well microtiter plates (Dynex Technologies, Chan-
tilly, VA) at 10 ␮g/mL. After washing with PBS, plates were blocked for
Five-micrometer cryosections of mouse spleen were stained with 7-amino-
1 hour with 0.5% bovine serum albumin in PBS at room temperature.
4-methylcoumarin-3-acetic acid–anti-mouse IgM (Vector Laboratories,
Mouse spleen cells (3 ⫻ 106 cells/well) or kidney suspension cells
Burlingame, CA), FITC-anti-mouse Thy1 (Pharmingen), and MOMA-1
(5 ⫻ 106 cells/well) in RPMI 1640 media with 10% fetal calf serum
mAb followed by horse anti–rat IgG/biotin and Cy3-Streptavidin (Jackson
Immunoresearch, West Grove, PA). Fluorescently labeled sections were were added into the first row of the plates and serially diluted at 1:2.
imaged on an Olympus BX-51 microscope (Olympus USA, Center Valley, Plates were cultured overnight at 37°C, cells were aspirated, and plates
PA) equipped with filter cubes for DAPI/AMCA, FITC/Cy2 and TRITC/ were washed 6 times followed by incubation with goat anti-mouse IgG
Cy3. Twelve-bit monochrome images were acquired with a 10⫻ objective (Southern Biotechnology Associates) at 1 ␮g/mL for 2 hours at 37°C.
using a Hamamatsu ORCA CCD camera (Olympus USA) driven by The plates were then washed 6 times with 0.05% Tween 20 in PBS.
Meta-Morph version 6.1 imaging software (Universal Imaging, Downing- Streptavidin-alkaline phosphatase was added to each well at 1:2000, and
town, PA). Image files were transferred to Adobe Photoshop CS2 (Adobe, plates were incubated at room temperature for 1 hour. After wash, the
Mountain View, CA), adjusted for contrast, and merged to create 24-bit plates were incubated with a mixture of 2-amino-2-methyl-1-propanol/5-
color images. bromo-4-chloro-3 indolyl phosphate) and 0.6% agarose gel overnight at
Formalin-fixed, paraffin-embedded cynomolgus monkey tissues were 4°C. Anti-ds/ssDNA IgG antibody-forming cells were enumerated as
sectioned at 4 to 6 ␮m and processed by standard techniques for blue spots using an inverted microscope and recorded as the number of
immunohistochemistry. Immunohistochemistry for B cells was accom- antibody-forming cells per spleen or per kidney.
3962 LIN et al BLOOD, 1 DECEMBER 2007 䡠 VOLUME 110, NUMBER 12

Figure 2. Kinetics and comparative mouse B-cell


reduction after treatment with anti-BR3 mAb, BR3-Fc,
or anti-CD20 mAb. Mice ( ⱖ 5/group) treated with anti-
BR3 mAb (200 ␮g/week), BR3-Fc (150 ␮g ⫻ 3/week), or
anti-CD20 mAb (200 ␮g/week) were analyzed at different
times, with B-cell numbers and subsets evaluated by flow
cytometry. Anti-BR3 treatment reduced blood B cells
(expressed as percentage of baseline absolute cell counts)
faster and more profoundly than BR3-Fc but slower than
anti-CD20 treatment (A). By day 15 of treatment, the
extent of B-cell reduction in the lymph node in response
to anti-BR3 treatment appeared greater than that result-
ing from anti-CD20 mAb treatment (B). B-cell subsets
were quantified using similar definitions as in Figure 1.
Spleen and bone marrow plasma cells were defined as
CD138⫹B220lo/⫺. Analysis of different B-cell subsets after
8 weeks of continuous treatment (200 ␮g/week anti-BR3
and anti-CD20; 150 ␮g ⫻ 3/week BR3-Fc) shows signifi-
cantly higher MZ B-cell, spleen, and BM plasma cell
reduction in anti-BR3 mAb compared with anti-CD20
mAb treatment (C). Serum IgM and IgG1 were measured
by ELISA after 3 months of continuous treatment (D).
These data are representative of 3 independent experi-
ments. Data are shown as mean plus or minus standard
error. Statistical significance compared with the control
treated group is indicated with an asterisk. When 2
treatment groups are significantly different from each
other, the P value is displayed.

confirmed by more frequent and longer-term treatments (data not


Results shown).

Anti-BR3 mAb treatment resulted in greater mouse B-cell Anti-BR3 treatment reduced mouse B cells more slowly but
reduction than BR3-Fc more profoundly in certain subsets than anti-CD20

Antibodies against mouse and human BR3 were generated by both To compare the effects of anti-BR3 and anti-CD20 mAbs on B-cell
classic hybridoma and phage display technologies and selected depletion, we used human CD20BACTg mice.13 Analysis of the
based on their ability to bind BR3 with high affinity, block BAFF kinetics of blood B-cell depletion with anti-BR3 revealed a 75%
binding to BR3, and prevent BAFF-mediated B-cell survival and reduction within 1 day and 99% over 8 days (Figure 2A). In
proliferation in B-cell cultures.47,49 A phage-derived antibody contrast, treatment with anti-CD20 mAb was faster at reducing
(CB2) was selected for further in vivo analysis.47 Fifteen days after blood B cells (75% reduction within 1 hour and 99% in 1 day),
in vivo administration of anti-BR3 (CB2) mAb in BALB/c mice, whereas the effect of BR3-Fc was slower than both anti-BR3 and
B cells were reduced in all lymphoid organs tested (Figure 1A-C). anti-CD20 (Figures 2A, S2). In lymph nodes, B-cell reduction
In blood, splenic follicles (FO; CD23⫹CD21int), splenic MZ achieved by anti-BR3 mAb occurred at a rate slower than that
(CD23loCD21hi), and lymph nodes, treatment with anti-BR3 mAb induced by anti-CD20 but became superior in magnitude by day 15
resulted in profound B-cell reduction (90%-99%) compared with (Figures 2B, S2).
isotype control-treated mice. In PP, FO cells (B220⫹CD38⫹) were To evaluate maximal B-cell reduction reached with these
reduced similarly to other recirculating blood, spleen, and lymph therapies in various B-cell subsets, we compared courses of
node compartments, whereas GC (B220⫹CD38lo) B cells were 8 weeks of continuous treatment with each reagent. Blood B cells
reduced to a lesser extent (Figure 1A,C). In the peritoneal cavity were reduced by 99% by both anti-CD20 and anti-BR3 mAbs and
(PEC), B2 cells (B220⫹CD23⫹) and B1 cells (B220⫹CD23⫺) were by 90%-95% in the BR3-Fc–treated mice (Figure 2C). In spleen
reduced by 90% and 70%–80%, respectively. Compared with the in and lymph nodes, FO cells were virtually eliminated by anti-BR3
vivo effects of BR3-Fc fusion protein, anti-BR3 mAb treatment mAb, whereas small residual subsets were seen after treatment with
caused a significantly higher level of B-cell reduction in blood, either anti-CD20 mAb (mostly MZ, few FO) or BR3-Fc (few FO).
spleen follicles, lymph nodes, and other compartments that contain Reduction of PP GC cells and spleen plasma cells were comparable
recirculating cells (PEC, PP follicles). In contrast, spleen marginal in mice treated with either anti-BR3 mAb or BR3-Fc (⬃75% and
zone B cells and plasma cells (PC) that do not recirculate and have 50%, respectively). In contrast, no significant reduction in PP GC
shown survival dependence on BAFF are reduced to similar levels or spleen plasma cells was observed after treatment with anti-CD20
(Figure 1A,C). These findings were confirmed by immunofluores- mAb, suggesting that decrease in these 2 subsets was probably due
cence on spleen sections, which shows complete depletion of FO to BAFF blockade.
and MZ regions in the anti-BR3 treated group with some residual The numbers of bone marrow (BM) PCs, which are enriched in
cells left in the follicles after BR3-Fc treatment (Figure 1B). Bright long-lived PCs, were not altered by long-term treatment with either
plasma cells (IgM⫹) in the red pulp are reduced in both BR3-Fc- anti-CD20 mAb or BR3-Fc. However, BM PCs showed a signifi-
and anti-BR3–treated groups. cant decrease (⬃50%) with anti-BR3 mAb treatment. These effects
Taken together, these data demonstrate that treatment with a on BM plasma cells translated into significant decreases in serum
depleting antagonistic anti-BR3 antibody results in superior B-cell IgM and IgG after anti-BR3 mAb treatment, whereas only IgG1
reduction compared with BR3-Fc. These results were further changed after 3 months of BR3-Fc treatment (Figure 2D and data
BLOOD, 1 DECEMBER 2007 䡠 VOLUME 110, NUMBER 12 IN VIVO B CELL EFFECTS OF ANTI-BR3 ANTIBODIES 3963

Figure 3. NHP B-cell reduction effects of anti-BR3


mAb, BR3-Fc, and anti-CD20 mAb. Comparison of
blood B cell numbers from cynomolgus monkeys treated
with anti-BR3 mAb (20 mg/kg/week: days 0-29, n ⫽ 16;
days 29-60, n ⫽ 10; terminal d29, n ⫽ 6), BR3-Fc (20 mg/
kg/week: days 0-29, n ⫽ 12; days 29-60, n ⫽ 4; terminal
d29, n ⫽ 8), anti-CD20 (2 ⫻ 50 mg/kg: days 0 and 14,
n ⫽ 4), and vehicle control (same numbers as active agent
groups) (A). Anti-BR3 appeared to reduce blood and
spleen B cells faster and more profoundly than BR3-Fc but
slower and less completely compared with anti-CD20
mAb. Naive lymph node B cells (CD20⫹CD21⫹CD27⫺)
were reduced similarly by anti-BR3 and anti-CD20 treat-
ments and less by BR3-Fc. Other lymph node B-cell
subsets—memory (CD20⫹CD21⫹CD27⫹), marginal zone
(CD20⫹CD21hiCD27⫺/⫹), germinal center (CD20⫹CD21⫺)
cells—were reduced similarly by all treatments (germinal
center cells were not assayed in the BR3-Fc-treated
experiment) (B). Statistical significance compared with the
control treated group is indicated with an asterisk. Because
treatment groups were in separate experiments, statistical
analysis was always performed against internal vehicle
control group. Treatment with anti-BR3 or BR3-Fc result in
spleen B-cell reduction measured by immunohistochemis-
try. B cells, blue (anti-CD20); T cells, orange (anti-CD3)
(C). See “Immunofluorescence and immunohistochemistry
staining” for image acquisition details.

not shown). Anti-CD20 mAb caused no reduction in serum differential sensitivity for BAFF blockade for these subsets in
immunoglobulin even after 1 year of continuous treatment.13 primates. Consistent with this lower primate sensitivity to BAFF
Because bone marrow plasma cells express low levels of BR3 but blockade compared with mice, serum immunoglobulins did not
not CD2050 in both mice and humans (Figure S3), anti-BR3 mAbs significantly change after the first 3 months of anti-BR3 mAb
may provide a therapeutic modality to target BM PCs. treatment (data not shown).
Compared with the anti-CD20 mAb and BR3-Fc studies,
The B-cell reduction profile of anti-BR3 was distinct from those anti-BR3 mAb was intermediate between the 2 in both kinetics and
of anti-CD20 and BR3-Fc in NHP degree of B-cell reduction in blood and spleen (Figure 3A-C).
BR3-Fc does not reduce blood B cells significantly in the first
B-cell reduction profiles elicited by BAFF-targeting reagents from 4 weeks, whereas anti-BR3 and anti-CD20 mAbs do, suggesting
mice and primates are different.17,19,20 Although mouse B cells are that cellular depletion, not survival blockade, is the main mecha-
exquisitely sensitive to BAFF targeting and more than 90% are nism responsible in this time frame for blood B-cell reduction.
eliminated within 2 weeks of treatment, NHP and human studies Reduction of lymph node B cells by anti-BR3 treatment seems to
have shown a maximum of only 50% overall reduction after 1 to be similar to that induced by anti-CD20 mAb (Figure 3B). These
6 months of treatment. This is due in part to different B-cell subset findings in NHP show clear differences of the BAFF/BR3 system
sensitivities for BAFF blockade (memory B cells, which comprise compared with rodents and have important consequences in
a larger part of the primate repertoire, appear less sensitive) but also guiding translatability of studies in rodents to treatment of human
to other unknown factors that might differ between rodents and diseases.
primates. To evaluate and help translate the anti-BR3 findings from
mouse to humans, we performed an NHP study in cynomolgus Anti-BR3 reduced some B-cell subsets more profoundly than
monkeys with a humanized antagonistic anti-BR3 antibody (h9.1). BR3-Fc in the NZB/W lupus-prone mouse strain
This clone is a surrogate of the mouse antibody (high affinity for
human and cynomolgus monkey BR3, BAFF-blocking in vitro To examine the effects of anti-BR3 immunotherapy, we evaluated
cellular assays, capable of ADCC, data not shown) while binding a the potency on autoimmune B-cell reduction of this dual targeting
different epitope on BR3.47 Monkeys were treated with anti-BR3, agent after treatment in the lupus-prone NZB/W mouse strain.
BR3-Fc, or anti-CD20 (in parallel experiments), and their B cells Seven- to 8-month-old NZB/W mice with established proteinuria
were compared with a control group. In contrast to mice, in which (30-100 mg/dL) were treated with control, anti-BR3 mAb, or
99% of blood B cells were eliminated within 1 to 2 weeks after BR3-Fc continuously for up to 5.5 months. Treatment with
anti-BR3 mAb treatment, blood B cells in cynomolgus monkeys anti-BR3 mAb decreased total B cells in blood and spleen to a
decreased by approximately 50% after 2 weeks (Figure 3A). greater extent compared with BR3-Fc (Figure S5). Although
Maximum blood B-cell reduction of approximately 75% was overall B-cell depletion in diseased NZB/W mice was efficient, the
reached after 1 month and remained near this level for the rest of reduction was less than that observed in normal healthy mice used
the treatment. Lymphoid organ analysis at a 4-week timepoint as control group in the same experiment (data not shown). As
using flow cytometry and immunohistochemistry showed that in early as 2 weeks (data not shown), and maximal at 6 weeks,
spleen and lymph nodes, B cells were decreased by approximately anti-BR3 mAb and BR3-Fc treatments decreased spleen anti-
80% (Figure 3B,C). Reduction of naive B cells in blood and lymph DNA-producing plasma cells, total plasma cells, and kidney-
nodes (both FO and MZ) was faster and more profound compared infiltrating B and T cells but left naive T cells unchanged (Figure
with memory B cells (Figure 3B and data not shown), suggesting a 4A). Spleen germinal center B cells and noncycling IgG1
3964 LIN et al BLOOD, 1 DECEMBER 2007 䡠 VOLUME 110, NUMBER 12

Figure 4. Anti-BR3 mAb decreases pathogenic B-cell


subsets in the NZB/W F1 SLE mouse model. Therapeu-
tic treatment of proteinuric NZB/W F1 mice (15/group) for
6 weeks with anti-BR3 mAb (600 ␮g/week) or BR3-Fc
(300 ␮g ⫻ 3/week) reduced splenic anti-DNA-producing
cells, kidney-infiltrating B cells (B220⫹), and T cells (CD3⫹)
but not naive spleen T cells (A). Splenic germinal center B
cells (B220⫹CD38lo) were decreased only by anti-BR3 mAb
treatment, whereas plasma cells (CD138⫹B220lo) were de-
creased by both treatments (B). After 1 week, anti-BR3
mAb and BR3-Fc treatment showed significant reduction
in both cycling and noncycling splenic B cells (B220⫹)
and IgG1 plasma cells (cytoplasmic IgG1⫹CD138⫹), but
only anti-BR3 mAb treatment resulted in significant reduc-
tion in IgM plasma cells (cytoplasmic IgM⫹CD138⫹) (C).
Statistical significance compared with the control treated
group is indicated with an asterisk. When 2 treatment
groups are significantly different from each other, the P
value is displayed.

plasma cells were reduced more with anti-BR3 mAb than Fc-mediated B-cell depletion and survival blockade contributed
BR3-Fc treatment (Figure 4B,C; detailed below). to the mechanism of anti-BR3 mAb action
To evaluate the impact of anti-BR3 mAb or BR3-Fc treat-
To further dissect in vivo the mechanisms by which anti-BR3
ment on cycling versus noncycling B-cell compartments, we
(CB2) mAb mediates B-cell reduction, we compared the wild-type
performed BrdU pulse experiments. In contrast to cyclophospha-
mAb with a D265A/N297A anti-BR3 mutant mAb incapable of Fc
mide (Cytoxan), which reduces mainly cycling cells, both
receptor binding and ADCC but still capable of BAFF/BR3
therapies decreased cycling and noncycling B-cell compart-
blockade.51 This anti-BR3 Fc mutant molecule had the same
ments effectively after 11 days (Figures 4C, S5). Likewise, both
affinity for BR3 and blocked BAFF-BR3 indistinguishably from
cycling and noncycling plasma cells were reduced; the isotype- the wild-type molecule (data not shown). We further compared the
switched plasma cells were more profoundly decreased than effects due to treatment with a nonblocking anti-BR3 mAb (PIH11)
IgM-positive plasma cells after therapy. Trends toward greater capable of inducing ADCC, but that does not prevent BAFF/BR3,
effects from anti-BR3 mAb compared with BR3-Fc treatment whereas a final comparison group was treated with BR3-Fc, which
were observed in all compartments, but statistical significance blocks BAFF binding to all its receptors without any B-cell-
between the 2 groups was reached only for IgG1 plasma cells. directed ADCC. One day after therapeutic administration, both
Effects of treatment on the plasma cell compartment were blocking and nonblocking anti-BR3 mAbs showed significant
paralleled by decreases in total serum IgM and IgG isotypes, as peripheral B-cell reduction, whereas the anti-BR3 mutant mAb and
well as of IgG anti-DNA antibodies (data not shown). In all BR3-Fc did not (Figure 5A). These data indicate that early
these readouts, anti-BR3 mAb was superior to BR3-Fc, suggest- peripheral B-cell reduction appeared to result solely from Fc-
ing that, similar to normal mice, effects on the autoimmune mediated mechanisms. After 6 days of treatment, a greater degree
plasma cell compartment are more pronounced with anti-BR3 of peripheral B-cell reduction was observed in mice treated with
mAb than BR3-Fc. wild-type anti-BR3 (CB2) mAb than in all other groups. Hence,
This series of experiments showed that anti-BR3 mAb reduced both Fc-mediated cell killing and BAFF/BR3 survival blockade
certain subsets (GC B cells, IgG1 plasma cells) more profoundly function in vivo and contribute to B-cell reduction.
than BR3-Fc, although in all other B-cell subsets, the 2 therapies For spleen and lymph node FO B cells, the best reduction was
have similar effects. again achieved with the blocking anti-BR3 (CB2) mAb, whereas

Figure 5. Anti-BR3 combines 2 mechanisms of action. BALB/c mice (5/group) were treated with the wild-type BAFF-blocking anti-BR3 mAb (CB2), anti-BR3 Fc mutant mAb
(CB2-DANA), anti-BR3 nonblocking mAb (PIH11), or BR3-Fc and evaluated for B-cell reduction at days 1 and 6. In blood at day 1, only treatments that had the ability to engage
Fc␥ receptors and induce ADCC showed B-cell reduction, whereas at day 6, ADCC and survival blockade appear to combine for maximal effects (A). In spleen FO cells, both
mechanisms appeared to contribute to B-cell reduction, whereas in MZ, the dominant mechanism appears to be BAFF/BR3-dependent survival blockade. At day 6, the spleen
plasma cell compartment was reduced only by ADCC competent molecules (B). Statistical significance compared with the control-treated group is indicated with an asterisk; P
values between groups are shown numerically.
BLOOD, 1 DECEMBER 2007 䡠 VOLUME 110, NUMBER 12 IN VIVO B CELL EFFECTS OF ANTI-BR3 ANTIBODIES 3965

all other groups showed lower but significant decreases in B-cell Because the primate immune system is populated at steady state
populations (Figure 5B). In contrast, the MZ compartment, which with a much larger proportion of memory, germinal center, and
is extremely BAFF dependent for survival but does not recirculate, plasma cells, compared with that in pathogen-free mice, pri-
demonstrated comparable degrees of B-cell reduction with anti- mates could be expected to be less affected by BAFF blockade
BR3 (CB2) mAb, mutant anti-BR3 (mutant CB2), and BR3-Fc therapy than mice. Supporting this hypothesis, B-cell reduction
(Figure 5B). That BAFF blockade represents a major mechanism of after treatment with anti-BR3 mAb in lupus-prone mice, which
B-cell reduction by anti-BR3 (CB2) mAb is further supported by have an activated immune system, is inferior to that seen in
the lesser reduction in MZ B cells by a nonblocking anti-BR3 normal BALB/c, C57BL/6, or FvB mice. In addition, unknown
(PIH11) mAb. primate B-cell survival characteristics could also contribute to
Finally, at day 6, spleen plasma cells were decreased by this apparent discrepancy between species. Initial results from
treatment with depleting blocking and nonblocking anti-BR3 mice deficient in members of the BAFF/BR3-induced alterna-
(CB2 and PIH11) but not by mutant anti-BR3 or BR3-Fc (Figure tive nuclear factor ␬B (NF-␬B) 2 pathway suggested that this
5B). This suggests that Fc-mediated depletion functions to pathway was indispensable for B-cell development and survival.
decrease tissue-resident plasma cells. Because longer treatments However, more recent studies have demonstrated that the classic
with BR3-Fc reduce this compartment in the spleen (Figures and alternative NF-␬B pathways can replace each other in
1C,2C) but not in bone marrow, Fc-mediated cell depletion certain conditions during B-cell ontogeny.53,54 Considering the
confers additional qualitative properties distinct from BAFF large number of cues feeding into these pathways during B-cell
survival blockade. development (eg, BCR, BR3, CD40, TLR, interleukins), it is
These data, taken together, demonstrate that a blocking anti- possible that during evolution, mice and primates developed
BR3 antibody reaches maximal efficiency to reduce B cells by different optimal combinations of survival and maturation
combining Fc-mediated cell killing with survival blockade of the signals. Further research in dissecting the developmental cues in
BAFF/BR3 pathway. various B-cell subsets in mice versus primates should shed light
on this question.
Another aspect of therapeutical targeting relates to the B-cell
subsets that are affected differentially with various strategies.
Discussion
Clinical and preclinical data shows that marginal zone B cells, as
In an attempt to create a therapeutic strategy combining B-cell well as memory, germinal center, and plasma cells, are deregulated
depletion with survival factor blockade, we generated anti-BR3 in human rheumatologic disorders. Marginal zone B cells associ-
mAbs with dual attributes—depletion and blockade of survival ated with high levels of circulating BAFF have been described in
signals. In both mice and primates, these antibodies were shown both human Sjögren syndrome salivary gland infiltrates as well as
to use a dual mechanism in reducing different B-cell subsets. In in a Sjögren syndrome mouse model.28,55 Likewise, subsets of
mice, the effect of anti-BR3 was slower than anti-CD20, faster patients with memory B cells, germinal center, and plasma cells
than BR3-Fc, and ultimately resulted in a maximal B-cell with different organization patterns in the joint have been described
reduction profile superior to anti-CD20 in particular B-cell in patients with rheumatoid arthritis.56 Recent data from arthritis
subsets. This was evident in subsets that are relatively resistant patients treated with anti-CD20 therapy show that B-cell depletion
to anti-CD20 depletion (eg, spleen marginal zone, Peyer patch in the bone marrow and joints is incomplete, although the
germinal centers, spleen, and bone marrow plasma cells). It is correlation of this finding with clinical response has not been
important to mention that although these conclusions reflect studied yet.57,58 A more direct parallel between incomplete malig-
comparisons between representative anti-BR3 and anti-CD20 nant B-cell depletion by immunotherapy and clinical relapse is
mAbs, other specific antibodies against different CD20 epitopes better understood in NHL.5 In this light, for clinical benefit,
and with additional or enhanced depleting properties could different human diseases with B-cell deregulation could require
behave somewhat differently. In NHP, similar to mice, anti-BR3 distinct levels of B-cell reduction better achieved through an
mAb B-cell depletion was faster than BR3-Fc and slower than appropriate but distinct B-cell immunotherapy strategy.
anti-CD20. However, the maximal B-cell reduction obtained Regardless of B-cell depletion level, because of the demon-
after 1 month of treatment was approximately 80% in certain strated dual mechanism of action of anti-BR3 mAbs, surviving
subsets (naive, follicular, and marginal zone) and less in others residual B cells will be in an environment without a functional
(memory, germinal center, and plasma cells). This reduction was BAFF/BR3 pathway. This is important for potential therapy
superior to previously reported anti-BAFF reagents (anti-BAFF because both in oncology (CLL, NHL) and immunology (RA, SLE,
antibodies, BR3-Fc, TACI-Fc), showing that anti-BR3 mAb, in Sjögren syndrome), increased BAFF levels signaling through its
addition to blocking survival, is also depleting B cells in NHP. receptors contribute to pathogenic B-cell survival.59,60
Compared with anti-CD20, which entirely depletes NHP blood BR3 was expressed at different levels on the surface of both the
and spleen B cells and incompletely depletes lymph node B malignant and normal residual B cells in a series of 16 patients with
cells, anti-BR3 B-cell reductions appear less in blood and spleen CLL who varied widely with respect to CD20 expression (3/16
and comparable in lymph nodes.52 The faster kinetics of B-cell were negative as evaluated by flow cytometry; Figure S6). It was
reduction of anti-CD20 is probably due to the higher density of suggested that some of the in vitro BAFF survival effects on CLL B
CD20 surface expression compared with BR3 as well as capping cells result through non-BR3 receptor (TACI) signaling and classic
differences between these molecules after mAb binding (data NF-␬B activation.59 However, a functional BR3 survival pathway
not shown). Although anti-CD20 depletion translates relatively does exist in CLL B cells, and its in vivo inhibition could be
well from rodents to primates, blockade of the BAFF/BR3 beneficial in addition to the anti-BR3 mAb cellular depletion
system does not. It is likely that this is due in part to a reduced mechanism. Compared with normal B cells, CLL B cells have
dependence for BAFF survival signals of memory, germinal lower levels of CD20 expression, which probably contributes to the
center B cells, and plasma cells compared with naive B cells. relative lower efficacy of anti-CD20 immunotherapy in CLL
3966 LIN et al BLOOD, 1 DECEMBER 2007 䡠 VOLUME 110, NUMBER 12

compared with NHL.61 Patients with negligible CD20 on CLL large number of studies, Wesley Cheng and Ronald Ferrando for
B cells that express significant BR3 constitute prime candidates for immunofluorescence, and the development sciences FACS labora-
potential therapeutic benefit from anti-BR3 treatment. It is impor- tory for the NHP FACS.
tant to point out that our data show properties of anti-BR3 mAb
largely in normal and mouse autoimmune B cells. Additional
experiments will be required to evaluate this therapy in the context
of malignant B cells. Authorship
Finally, reduction of BM plasma cells after anti-BR3 mAb
treatment in mice parallels the synergistic effect seen with combin- Contribution: W.Y.L, Q.G., D.S., Z.L., W.P.L., K.L., S.Y.,
ing anti-CD20 mAb and BR3-Fc.13 If this effect translates even L.E.D., Q.O., S.Y., E.S., J.S., M.B., M.A.S., and F.M. performed
partially to humans, it should have therapeutic value in subsets of research, collected, analyzed and interpreted data; K.H., M.L.,
autoimmune patients with pathogenic long-lived autoantibody S.I., J.B., D.D., and T.G. analyzed and interpreted data; CW.L.,
components. Future clinical trials in both oncology and immunol- J.F., J.S.T., C.A., and A.E. contributed vital new reagents; T.G.,
ogy will test the translatability and therapeutic value of B-cell M.B., M.A.S., and F.M. designed research; and FM drafted the
reduction through anti-BR3 mAbs in human disease. manuscript.
Conflict-of-interest disclosure: The authors are employees of
Genentech, with the exception of J.S.T. and C.A., who are
Acknowledgments employees of Biogen-IDEC.
Correspondence: Flavius Martin, Department of Immunology,
We acknowledge the hard work and dedication of the in vivo Genentech, Inc, 1 DNA Way, MS34, 12-286, South San Francisco,
immunology group in dosing, tracking down, and analyzing a very CA 94080; e-mail: flavius@gene.com.

References
1. Jacobsen E, Freedman A. B-cell purging in au- depletion during CD20 immunotherapy. J Exp 27. Cambridge G, Stohl W, Leandro MJ, et al. Circu-
tologous stem-cell transplantation for non- Med. 2006;203:743-753. lating levels of B lymphocyte stimulator in pa-
Hodgkin lymphoma. Lancet Oncol. 2004;5:711- 15. Teeling JL, French RR, Cragg MS, et al. Charac- tients with rheumatoid arthritis following rituximab
717. terization of new human CD20 monoclonal anti- treatment: relationships with B cell depletion, cir-
2. Martin F, Chan AC. B cell immunobiology in dis- bodies with potent cytolytic activity against non- culating antibodies, and clinical relapse. Arthritis
ease: evolving concepts from the clinic. Annu Rev Hodgkin lymphomas. Blood. 2004;104:1793- Rheum. 2006;54:723-732.
Immunol. 2006;24:467-496. 1800. 28. Lavie F, Miceli-Richard C, Ittah M, et al. Increase
3. Browning JL. B cells move to centre stage: novel 16. Hamaguchi Y, Uchida J, Cain DW, et al. The peri- of B cell-activating factor of the TNF family
opportunities for autoimmune disease treatment. toneal cavity provides a protective niche for B1 (BAFF) after rituximab: insights into a new regu-
Nat Rev Drug Discov. 2006;5:564-576. and conventional B lymphocytes during anti- lating system of BAFF production. Ann Rheum
CD20 immunotherapy in mice. J Immunol. 2005; Dis. 2007;66:700-703.
4. Edwards JC, Cambridge G. B-cell targeting in
174:4389-4399. 29. Toubi E, Kessel A, Slobodin G, et al. Changes in
rheumatoid arthritis and other autoimmune dis-
eases. Nat Rev Immunol. 2006;6:394-403. 17. Kalled SL. The role of BAFF in immune function macrophage function after rituximab treatment in
and implications for autoimmunity. Immunol Rev. patients with rheumatoid arthritis. Ann Rheum
5. Maloney DG. Immunotherapy for non-Hodgkin’s Dis. 2007;66:818-820.
2005;204:43-54.
lymphoma: monoclonal antibodies and vaccines.
J Clin Oncol. 2005;23:6421-6428. 18. Mackay F, Schneider P, Rennert P, Browning J. 30. Vallerskog T, Heimburger M, Gunnarsson I, et al.
BAFF AND APRIL: a tutorial on B cell survival. Differential effects on BAFF and APRIL levels in
6. Cvetković RS, Perry CM. Rituximab: a review of rituximab-treated patients with systemic lupus
Annu Rev Immunol. 2003;21:231-264.
its use in non-Hodgkin’s lymphoma and chronic erythematosus and rheumatoid arthritis. Arthritis
lymphocytic leukaemia. Drugs. 2006;66:791-820. 19. Vugmeyster Y, Seshasayee D, Chang W, et al. A
soluble BAFF antagonist, BR3-Fc, decreases pe- Res Ther. 2006;8:R167.
7. Edwards JC, Szczepanski L, Szechinski J, et al. ripheral blood B cells and lymphoid tissue mar- 31. Shields RL, Lai J, Keck R, et al. Lack of fucose on
Efficacy of B-cell-targeted therapy with rituximab ginal zone and follicular B cells in cynomolgus human IgG1 N-linked oligosaccharide improves
in patients with rheumatoid arthritis. N Engl monkeys. Am J Pathol. 2006;168:476-489. binding to human Fc␥ RIII and antibody-depen-
J Med. 2004;350:2572-2581. dent cellular toxicity. J Biol Chem. 2002;277:
20. Halpern WG, Lappin P, Zanardi T, et al. Chronic
8. Ahmed AR, Spigelman Z, Cavacini LA, Posner administration of belimumab, a BLyS antagonist, 26733-26740.
MR. Treatment of pemphigus vulgaris with ritux- decreases tissue and peripheral blood B-lympho- 32. Presta LG, Shields RL, Namenuk AK, Hong K,
imab and intravenous immune globulin. N Engl cyte populations in cynomolgus monkeys: phar- Meng YG. Engineering therapeutic antibodies for
J Med. 2006;355:1772-1779. macokinetic, pharmacodynamic, and toxicologic improved function. Biochem Soc Trans. 2002;30:
9. Silverman GJ. Therapeutic B cell depletion and effects. Toxicol Sci. 2006;91:586-599. 487-490.
regeneration in rheumatoid arthritis: emerging 21. Ding C, Jones G. Belimumab Human Genome 33. Idusogie EE, Wong PY, Presta LG, et al. Engi-
patterns and paradigms. Arthritis Rheum. 2006; Sciences/Cambridge Antibody Technology/ neered antibodies with increased activity to re-
54:2356-2367. GlaxoSmithKline. Curr Opin Investig Drugs. cruit complement. J Immunol. 2001;166:2571-
10. Martin F, Chan AC. Pathogenic roles of B cells in 2006;7:464-472. 2575.
human autoimmunity; insights from the clinic. Im- 22. Gross JA, Dillon SR, Mudri S, et al. TACI-Ig neu- 34. Gillies SD, Lan Y, Williams S, et al. An anti-CD20-
munity. 2004;20:517-527. tralizes molecules critical for B cell development IL-2 immunocytokine is highly efficacious in a
11. Clynes RA, Towers TL, Presta LG, Ravetch JV. and autoimmune disease. Impaired B cell matu- SCID mouse model of established human B lym-
Inhibitory Fc receptors modulate in vivo cytoxicity ration in mice lacking BLyS. Immunity. 2001;15: phoma. Blood. 2005;105:3972-3978.
against tumor targets. Nat Med. 2000;6:443-446. 289-302.
35. Adamson PJ, Millard DJ, Hohmann AW, et al. Im-
12. Uchida J, Hamaguchi Y, Oliver JA, et al. The in- 23. Thompson JS, Bixler SA, Qian F, et al. BAFF-R, a proved antigen binding by a CD20-specific single-
nate mononuclear phagocyte network depletes B newly identified TNF receptor that specifically in- chain antibody fragment with a mutation in
lymphocytes through Fc receptor-dependent teracts with BAFF. Science. 2001;293:2108-2111. CDRH1. Mol Immunol. 2006;43:550-558.
mechanisms during anti-CD20 antibody immuno- 24. Cohen SB. Updates from B cell trials: efficacy. 36. Yazawa N, Hamaguchi Y, Poe JC, Tedder TF. Im-
therapy. J Exp Med. 2004;199:1659-1669. J Rheumatol Suppl. 2006;77:12-17. munotherapy using unconjugated CD19 mono-
13. Gong Q, Ou Q, Ye S, et al. Importance of cellular 25. Looney RJ. B-cell-targeted therapy for rheuma- clonal antibodies in animal models for B lympho-
microenvironment and circulatory dynamics in B toid arthritis: an update on the evidence. Drugs. cyte malignancies and autoimmune disease.
cell immunotherapy. J Immunol. 2005;174:817- 2006;66:625-639. Proc Natl Acad Sci U S A. 2005;102:15178-
826. 26. Lesley R, Xu Y, Kalled SL, et al. Reduced com- 15183.
14. Hamaguchi Y, Xiu Y, Komura K, Nimmerjahn F, petitiveness of autoantigen-engaged B cells due 37. Steinfeld SD, Youinou P. Epratuzumab (human-
Tedder TF. Antibody isotype-specific engagement to increased dependence on BAFF. Immunity. ised anti-CD22 antibody) in autoimmune dis-
of Fcgamma receptors regulates B lymphocyte 2004;20:441-453. eases. Expert Opin Biol Ther. 2006;6:943-949.
BLOOD, 1 DECEMBER 2007 䡠 VOLUME 110, NUMBER 12 IN VIVO B CELL EFFECTS OF ANTI-BR3 ANTIBODIES 3967

38. Strauss SJ, Morschhauser F, Rech J, et al. Multi- recombinant interleukin-2 for rituximab-refractory motes B cell proliferation upon activation. Immu-
center phase II trial of immunotherapy with the indolent non-Hodgkin’s lymphoma. Clin Cancer nity. 2006;24:729-739.
humanized anti-CD22 antibody, epratuzumab, in Res. 2006;12:7046-7053. 54. Enzler T, Bonizzi G, Silverman GJ, et al. Alterna-
combination with rituximab, in refractory or recur- 46. Kimby E. Beyond immunochemotherapy: combi- tive and classical NF-kappaB signaling retain au-
rent non-Hodgkin’s lymphoma. J Clin Oncol. nations of rituximab with cytokines interferon- toreactive B cells in the splenic marginal zone
2006;24:3880-3886. alpha2a and granulocyte colony stimulating factor and result in lupus-like disease. Immunity. 2006;
39. Tedder TF, Baras A, Xiu Y. Fcgamma receptor- [published erratum appears in Semin Oncol. 25:403-415.
dependent effector mechanisms regulate CD19 2002;29:609]. Semin Oncol. 2002;29:7-10. 55. Groom J, Kalled SL, Cutler AH, et al. Association
and CD20 antibody immunotherapies for B lym-
47. Lee CV, Hymowitz SG, Wallweber HJ, et al. Syn- of BAFF/BLyS overexpression and altered B cell
phocyte malignancies and autoimmunity.
thetic anti-BR3 antibodies that mimic BAFF bind- differentiation with Sjögren’s syndrome. J Clin
Springer Semin Immunopathol. 2006;28:351-364.
ing and target both human and murine B cells. Invest. 2002;109:59-68.
40. Wu AM, Senter PD. Arming antibodies: prospects Blood. 2006;108:3103-3111. 56. Weyand CM, Kang YM, Kurtin PJ, Goronzy JJ.
and challenges for immunoconjugates. Nat Bio-
48. Ng LG, Sutherland AP, Newton R, et al. B cell- The power of the third dimension: tissue architec-
technol. 2005;23:1137-1146.
activating factor belonging to the TNF family ture and autoimmunity in rheumatoid arthritis.
41. Dunussi-Joannopoulos K, Hancock GE, Kunz A, (BAFF)-R is the principal BAFF receptor facilitat- Curr Opin Rheumatol. 2003;15:259-266.
et al. B-cell depletion inhibits arthritis in a colla- ing BAFF costimulation of circulating T and B
gen-induced arthritis (CIA) model, but does not 57. Vos K, Thurlings RM, Wijbrandts CA, et al. Early
cells. J Immunol. 2004;173:807-817. effects of rituximab on the synovial cell infiltrate in
adversely affect humoral responses in a respira-
tory syncytial virus (RSV) vaccination model. 49. Avery DT, Kalled SL, Ellyard JI, et al. BAFF selec- patients with rheumatoid arthritis. Arthritis
Blood. 2005;106:2235-2243. tively enhances the survival of plasmablasts gen- Rheum. 2007;56:772-778.
erated from human memory B cells. J Clin Invest. 58. Leandro MJ, Cooper N, Cambridge G, Ehrenstein
42. Polson AG, Yu SF, Elkins K, et al. Antibody-drug 2003;112:286-297.
conjugates targeted to CD79 for the treatment of MR, Edwards JC. Bone marrow B-lineage cells in
non-Hodgkin’s lymphoma. Blood. 2007;110:616- 50. Harada H, Kawano MM, Huang N, et al. Pheno- patients with rheumatoid arthritis following ritux-
623. typic difference of normal plasma cells from ma- imab therapy. Rheumatology (Oxford). 2007;46:
ture myeloma cells. Blood. 1993;81:2658-2663. 29-36.
43. Niitsu N, Hayama M, Okamoto M, et al. Phase I
study of rituximab-CHOP regimen in combination 51. Shields RL, Namenuk AK, Hong K, et al. High 59. Endo T, Nishio M, Enzler T, et al. BAFF and
with granulocyte colony-stimulating factor in pa- resolution mapping of the binding site on human APRIL support chronic lymphocytic leukemia B-
tients with follicular lymphoma. Clin Cancer Res. IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma cell survival through activation of the canonical
2004;10:4077-4082. RIII, and FcRn and design of IgG1 variants with NF-kappaB pathway. Blood. 2007;109:703-710.
improved binding to the Fc gamma R. J Biol 60. Becker-Merok A, Nikolaisen C, Nossent HC. B-
44. Niitsu N, Khori M, Hayama M, et al. Phase I/II
Chem. 2001;276:6591-6604. lymphocyte activating factor in systemic lupus
study of the rituximab-EPOCT regimen in combi-
nation with granulocyte colony-stimulating factor 52. Vugmeyster Y, Beyer J, Howell K, et al. Depletion erythematosus and rheumatoid arthritis in relation
in patients with relapsed or refractory follicular of B cells by a humanized anti-CD20 antibody to autoantibody levels, disease measures and
lymphoma including evaluation of its cardiotoxic- PRO70769 in Macaca fascicularis. J Immunother. time. Lupus. 2006;15:570-576.
ity using B-type natriuretic peptide and troponin T 2005;28:212-219. 61. Wierda WG, Kipps TJ, Keating MJ. Novel im-
levels. Clin Cancer Res. 2005;11:697-702. 53. Sasaki Y, Derudder E, Hobeika E, et al. Canonical mune-based treatment strategies for chronic lym-
45. Khan KD, Emmanouilides C, Benson DM Jr, et al. NF-kappaB activity, dispensable for B cell devel- phocytic leukemia. J Clin Oncol. 2005;23:6325-
A phase 2 study of rituximab in combination with opment, replaces BAFF-receptor signals and pro- 6332.

You might also like