Anda di halaman 1dari 12

0022-3565/09/3303-864–875$20.

00
THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS Vol. 330, No. 3
Copyright © 2009 by The American Society for Pharmacology and Experimental Therapeutics 153973/3502930
JPET 330:864–875, 2009 Printed in U.S.A.

The Binding Specificity and Selective Antagonism of


Vedolizumab, an Anti-␣4␤7 Integrin Therapeutic Antibody in
Development for Inflammatory Bowel Diseases

Dulce Soler, Tobias Chapman, Li-Li Yang, Tim Wyant, Robert Egan, and Eric R. Fedyk
Millennium Pharmaceuticals Inc., a Takeda Pharmaceuticals Company, Cambridge, Massachusetts
Received March 26, 2009; accepted June 8, 2009

ABSTRACT
Vedolizumab is a humanized monoclonal antibody that targets and basophils at lower levels; vedolizumab binds to memory
the ␣4␤7 integrin exclusively, and modulates inflammation in the CD4⫹ T and B lymphocytes with subnanomolar potency
gastrointestinal tract without inducing the systemic immuno- (EC50 ⫽ 0.3– 0.4 nM). The second determinant is binding spec-
suppression that characterizes anti-␣4 chain monoclonal anti- ificity; vedolizumab binds exclusively to the ␣4␤7 integrin, and
bodies, such as natalizumab. This unique pharmacologic profile not to the ␣4␤1 and ␣E␤7 integrins. The third determinant is
is largely attributable to four determinants. The first determinant selective antagonism; vedolizumab selectively inhibits adhe-
is the restriction of the expression of the ␣4␤7 integrin to sub- sion of ␣4␤7-expressing cells to mucosal addressin cell adhe-
sets of leukocytes. Vedolizumab does not bind to the majority sion molecule 1 (median inhibition concentration [IC50] ⫽ 0.02–
of memory CD4⫹ T lymphocytes (60%), neutrophils, and most 0.06 ␮g/ml) and fibronectin (IC50 ⫽ 0.02 ␮g/ml), but not
monocytes. The highest level of vedolizumab binding is to a vascular cell adhesion molecule 1. The fourth determinant is the
subset (⬃25%) of human peripheral blood memory CD4⫹ T gastrointestinal-specific tropism of the ␣4␤7 integrin function.
lymphocytes that include gut-homing interleukin 17 T-helper These pharmacologic properties of vedolizumab, in conjunction
lymphocytes. Vedolizumab also binds to eosinophils at high with the gastrointestinal tropism of ␣4␤7 integrin function, may
levels, and to naive T-helper lymphocytes, naive and memory ultimately confer an improved risk-to-benefit profile for patients
cytotoxic T lymphocytes, B lymphocytes, natural killer cells, with inflammatory bowel diseases.

The inflammatory bowel diseases (IBDs), ulcerative colitis these lymphocytes enter the gut are distinct from those in
(UC) and Crohn’s disease (CD), are chronic diseases of the other peripheral tissues, such as the skin and central ner-
gastrointestinal (GI) tract characterized by an exacerbated vous system (Butcher and Picker, 1996; Engelhardt et al.,
inflammatory cell infiltrate in the gut mucosal tissue (Xavier 1998; Engelhardt and Briskin, 2005; Salmi and Jalkanen,
and Podolsky, 2007). Multiple inflammatory cell types, in- 2005; Agace, 2006). The complex infiltration process in the GI
cluding neutrophils, macrophages, dendritic cells, and lym- tract requires the coordinated interaction of several adhesion
phocytes, participate in the pathogenesis of IBD, with lym- and signaling molecules on the surface of T lymphocytes
phocytes having a central role in the induction and (selectins, integrins, chemokine receptors) with their corre-
maintenance of the chronic inflammatory process in the lam- sponding ligands on the endothelium. The ␣4␤7 integrin me-
ina propria (Xavier and Podolsky, 2007). Infiltration of the GI diates the infiltration of the GI tract by memory T lympho-
tract by T lymphocytes is a well documented pathogenic cytes, binding to mucosal addressin cell adhesion molecule 1
mechanism of IBD, and the molecular mechanisms by which (MAdCAM-1) on endothelial cells, and blockade of this inter-
action provides efficacy in animal models of IBD (Hesterberg
This work was supported by Millennium Pharmaceuticals Inc. et al., 1996; Picarella et al., 1997), and in patients with UC
Some of these data were presented at the 2008 Advances in Inflammatory (Feagan et al., 2005) and CD (Feagan et al., 2008).
Bowel Diseases Crohn’s and Colitis Foundation’s Clinical and Research Con-
ference; 2008 Dec 4 –7; Hollywood, FL; and at the 4th Congress of the European The ␣4␤7 integrin is consequently an ideal therapeutic
Crohn’s and Colitis Organisation; 2009 Feb 5–7; Hamburg, Germany. target for IBD and is currently being targeted through three
Article, publication date, and citation information can be found at
http://jpet.aspetjournals.org. different strategies. Two of these strategies target either the
doi:10.1124/jpet.109.153973. ␣4 chain or the ␤7 chain. They are not specific for the ␣4␤7

ABBREVIATIONS: IBD, inflammatory bowel disease; BSA, bovine serum albumin; CD, Crohn’s disease; Fc, fragment crystallizable; GI, gastro-
intestinal; GMFI, geometric mean fluorescence intensity; mAb, monoclonal antibody; IL, interleukin; MAdCAM-1, mucosal vascular addressin cell
adhesion molecule 1; PBMC, peripheral blood mononuclear cell; PBS, phosphate-buffered saline; PML, progressive multifocal leukoencepha-
lopathy; Th17, T-helper 17; UC, ulcerative colitis; VCAM-1, vascular cell adhesion molecule 1.

864
Binding and Inhibitory Properties of Vedolizumab 865
integrin and bind to other integrins containing these chains, ing the CS-1 peptide, and unlabeled blocking antibodies to human
specifically the ␣4␤1 and ␣E␤7 integrins. The ␣4␤1 and ␣E␤7 VCAM-1, ␣4 and ␤1 were obtained from R&D Systems (Minneapolis,
integrins mediate effects within and outside the GI tract. The MN). A MAdCAM-1/mFc fusion protein was available in-house. Flu-
␣E␤7 integrin is postulated to locate and retain T lympho- orochrome-conjugated anti-mouse IgG was purchased from Jackson
cytes within the epithelium of numerous tissues by binding Immunoresearch Laboratories (West Grove, PA). Pharmlyse buffer
was obtained from BD Biosciences (San Jose, CA). Fluorochrome-
E-cadherin on the basolateral surface of epithelial cells
conjugated antibodies to human proteins were purchased from var-
(Kilshaw, 1999). The ␣4␤1 integrin mediates extravasation of ious sources: mouse antibodies to CD4 (L200), CD8 (SK1), CD14
lymphocytes, monocytes, and eosinophils into numerous (M␸P9), CD19 (HIB19), CD45R0 (UCHL1), CD49d (␣4; L25), CD56
types of tissues by binding to vascular cell adhesion molecule (NCAM16.2), and CD103 (␣E; Ber-ACT8) and a rat antibody to ␤7
1 (VCAM-1) expressed on the luminal surface of endothe- (FIB504) from BD Biosciences (San Jose, CA); mouse antibody to
lium, and to fibronectin within extracellular matrix (Gonzá- CD29 (␤1; MEM-101A) from eBioscience (San Diego, CA); mouse
lez-Amaro et al., 2005). Antagonizing the ␣4␤1 or ␣E␤7 inte- antibody to CD123 (AC145) from Miltenyi Biotech (Auburn, CA);
grin thus has a systemic effect. mouse antibodies to CD49d (␣4; 2B4), CD29 (␤1; P5D2), and VCAM-1
The humanized anti-␣4 antibody, natalizumab, elicits ef- (HAE-2Z) from R&D Systems. Unconjugated antibodies to the ␣4␤7
fects in numerous tissues, including leukocytosis (Ghosh et integrin, namely Act-1 (mouse) and vedolizumab (humanized Act-1),
and a humanized control antibody with a different antigen specificity
al., 2003; Sandborn et al., 2005; Targan et al., 2007), mobili-
but the same Fc IgG1 domain as vedolizumab, were available in-
zation of hematopoietic stem cells (Bonig et al., 2008; Zohren house, in addition to Alexa Fluor 647-conjugated vedolizumab or
et al., 2008), and inhibition of leukocyte trafficking into the biotin-Act-1. Primary antibodies for the human tissue cross-reactiv-
central nervous system (del Pilar Martin et al., 2008). In- ity investigation were rabbit anti-vedolizumab (generated in-house),
deed, antagonizing both the ␣4␤1 and ␣4␤7 integrins may anti-␤2-microglobulin (Dako North America, Inc., Carpinteria, CA),
explain efficacy in the central nervous system (Miller et al., and a negative control human IgG1 (Millipore Bioscience Research
2003) and the GI tract, respectively (Ghosh et al., 2003; Reagents, Temecula, CA).
Sandborn et al., 2005; Targan et al., 2007). However, admin- Cell Lines and Culture Media. The human B-cell lymphoma
istration of natalizumab is associated with systemic immu- cell line, RPMI8866 (stably expressing ␣4␤7), was a kind gift from Dr.
nosuppression; for example, increased incidence of the fatal David Erle (University of California, San Francisco). The human
B-cell lymphoma cell line, RAMOS (stably expressing ␣4␤1, origi-
infectious disease progressive multifocal leukoencephalopa-
nally sourced from the American Type Culture Collection, Manassas,
thy (PML) (Berger and Koralnik, 2005; Kleinschmidt-De-
VA), and ␣E␤7 L1.2 cell transfectants were generated in-house by use
Masters and Tyler, 2005; Langer-Gould et al., 2005; Ranso- of cDNA provided by Christina Parker and Michael Brenner
hoff, 2005; Van Assche et al., 2005). It is postulated that the (Brigham and Women’s Hospital, Boston, MA). Culture medium for
anti-inflammatory mechanism driving the efficacy of natali- RPMI8866 and RAMOS cell lines consisted of RPMI 1640 medium
zumab in multiple sclerosis may also predispose patients to supplemented with 1% penicillin/streptomycin and 1% L-glutamine
progressive multifocal leukoencephalopathy by decreasing (Invitrogen, Carlsbad, CA), and 10% US-defined fetal bovine serum
immunosurveillance in the central nervous system (Berger, (Hyclone Laboratories, Logan, UT). Culture medium for ␣E␤7 trans-
2006; Berger and Houff, 2006; Koralnik, 2006; Niino et al., fectants consisted of RPMI 1640 medium supplemented with 1%
2006; Stüve et al., 2006a,b; del Pilar Martin et al., 2008). penicillin/streptomycin, 1% sodium pyruvate, 1% L-glutamine, and 2
␮g/ml puromycin (Invitrogen), 0.1% ␤-mercaptoethanol (Sigma-Al-
A third strategy is exclusive targeting of the ␣4␤7 integrin,
drich, St. Louis, MO), and 10% US-defined fetal bovine serum (Hy-
which is used by vedolizumab (former versions known as
clone Laboratories).
MLN0002, MLN02, and LDP-02). Vedolizumab is a human- Immunohistochemistry of Normal Human Tissues. The
ized version of Act-1, a mouse antibody (Lazarovits et al., binding specificity of vedolizumab in 38 different types of normal
1984) that binds to a conformational epitope that is unique to human tissues (three independent donors per tissue type) was in-
the heterodimerization of the human ␣4 chain with the ␤7 vestigated by immunohistochemistry. The quality of these tissues
chain (Schweighoffer et al., 1993; Tidswell et al., 1997). Act-1 was verified by robust staining with a positive-control antibody
therefore binds specifically to the ␣4␤7 integrin, and admin- against ␤2-microglobulin (Dako North America, Inc.). Sections (5 ␮m)
istration to colitic cotton-top tamarins leads to the resolution were cut from fresh-frozen tissue samples embedded in OCT Com-
of disease (Hesterberg et al., 1996). Vedolizumab binds to the pound (Sakura Finetek USA, Inc., Torrance, CA) and fixed in acetone
for 10 min at room temperature. Just before staining, slides were
␣4␤7 integrin on peripheral blood lymphocytes and inhibits
fixed for 10 s in 10% neutral buffered formalin. Acetone/formalin-
adhesion of the lymphocyte to MAdCAM-1. Humanized Act-1
fixed cryosections were rinsed twice in phosphate-buffered saline
has demonstrated statistically significant efficacy in placebo- (PBS) and incubated for 20 min with a protein block (PBS; 0.5%
controlled phase 2 clinical trials of patients with moderately casein; 5% human gamma globulins; 0.02% goat IgG; 1 mg/ml heat-
active UC (Feagan et al., 2005) and in patients with moder- aggregated human IgG) designed to reduce nonspecific binding. Un-
ately active CD (Feagan et al., 2008). The enhanced specific- conjugated vedolizumab or a negative control human IgG1 (Millipore
ity of vedolizumab may ultimately confer an improved risk- Bioscience Research Reagents) were applied to sections at 2 or 20
to-benefit ratio for patients with IBD. The data reported ␮g/ml and incubated at room temperature for 1 h. The slides were
herein characterize the binding specificity and potency of then rinsed twice with PBS and an indirect immunoperoxidase pro-
vedolizumab and the associated selective antagonism of cedure was performed to detect these primary reagents. The second-
ary antibody, rabbit anti-vedolizumab, was then applied at 5 ␮g/ml
adhesion.
for 30 min and rinsed twice with PBS. Endogenous peroxidase was
blocked by incubating the slides for 5 min with the peroxidase solu-
tion provided in the Dako EnVision⫹ Kit and then rinsing twice with
Materials and Methods
PBS. The slides were then treated for 30 min with the peroxidase-
Proteins and Antibodies. Ninety-six-well plates coated with labeled goat anti-rabbit IgG polymer supplied in the Dako EnVi-
recombinant human VCAM-1/fragment crystallizable (Fc) chimera sion⫹ Kit, rinsed twice with PBS, and treated for 8 min with the
protein, an alternatively spliced form of human fibronectin contain- substrate-chromogen (DAB⫹) solution supplied in the Dako EnVi-
866 Soler et al.

sion⫹ Kit. All slides were rinsed with tap water, counterstained with ing buffer at 4°C for 30 min. After fixation and permeabilization,
hematoxylin, washed, “blued” in saturated lithium carbonate, cells were washed with permeabilization buffer (eBioscience) and
washed, dehydrated through alcohols, cleared in xylene, and cover- blocked with 10% normal rat serum at 4°C for 10 min. After the
slipped. Staining intensity was graded semiquantitatively by a blocking step, cells were stained with fluorochrome-conjugated anti-
board-certified anatomic pathologist. IL17A antibody (eBioscience) at 4°C for 30 min. Finally, cells were
Staining of Cell Lines and Whole Blood. For the staining of washed with permeabilization buffer and analyzed by flow cytometry
cell lines, cells were resuspended at 2 ⫻ 106/ml in FACS buffer (5% in a FACSCalibur with CellQuest Pro software. The memory Th17
fetal bovine serum and 0.05% sodium azide in D-PBS (Dulbecco’s population was examined for expression of the ␣4␤7 integrin and the
phosphate buffers saline without calcium and magnesium) (VWR, ␣4 and ␤7 chains. Some experiments were performed with isolated
West Chester, PA) and 200-␮l samples were stained with the appro- CD4⫹ memory T lymphocytes (Miltenyi Biotech) with very similar
priate monoclonal antibodies (see “Proteins and Antibodies”) at 4°C results to those with PBMCs.
for 30 min. Samples were washed with FACS buffer and analyzed by Adhesion Assays. For MAdCAM-1 and VCAM-1 experiments,
flow cytometry (FACSCalibur, BD). For the staining of human whole 96-well ELISA plates (Thermo Fisher Scientific, Waltham, MA) were
blood, 200-␮l samples from healthy human volunteers were stained prepared 24 h in advance by coating with 100 ␮l of 2 ␮g/ml recom-
with the appropriate monoclonal antibody at 4°C for 30 min. Red binant protein diluted in PBS at 4°C. For fibronectin experiments,
blood cells were lysed with BD FACS lysing solution, and samples commercially available precoated plates were used (see “Proteins and
then washed with FACS buffer and analyzed by flow cytometry. In Antibodies”). On the assay day, the coating solution was removed
all cases, antibodies were used at saturating concentrations and, in before blocking with blocking buffer [0.5% bovine serum albumin
many cases, up to four antibodies were used per sample. Appropriate (BSA), Sigma-Aldrich, in PBS] at 37°C (5% CO2/95% O2) for 1 h.
single- or two-color control stains were also performed. Experiments on low- and high-affinity binding to integrins were
Saturation and Competitive Binding Analyses. The potency performed in the absence or presence of Mn2⫹, respectively; Mn2⫹
of vedolizumab binding to human leukocytes was examined through was added to induce the high-affinity conformation of the integrin.
the generation of 1) antibody saturation binding curves with labeled Assay buffer for low-affinity experiments was RPMI 1640 medium,
vedolizumab or Act-1, and 2) antibody binding competition curves for 0.1% BSA, 10 mM HEPES, pH 7, and for high-affinity experiments
the competing binding of labeled antibody with unlabeled antibodies was 1 mM Mn2⫹, Dulbecco’s modified Eagle’s medium, 0.1% BSA, 10
(vedolizumab, Act-1, or isotype control IgG). Experiments were per- mM HEPES, pH 7. For dose-dependent antibody inhibition curves,
formed in 96-well v-bottom plates (Corning Inc., Corning, NY). For antibodies were diluted and cells resuspended in the indicated cor-
saturation binding experiments and determination of EC50 values, responding assay buffers for low- or high-affinity binding conditions.
100 ␮l of human peripheral blood from healthy human volunteers Premixed cell-antibody samples (100 ␮l at 2 ⫻ 106 cells/ml) were
was incubated with the labeled antibody at the indicated range of added to the protein-coated plates after removal of the blocking
concentrations in a final volume of 200 ␮l. In these saturation ex- buffer. Plates were incubated at 37°C (5% CO2/95% O2) for 1 h,
periments, at all concentrations tested, specific binding was demon- washed two to five times with PBS, and adhered cells were detected
strated by competition with 20-fold molar excess of unlabeled anti- by adding alamarBlue (Trek Diagnostic Systems, Cleveland, OH).
body. For binding competition experiments, 100 ␮l of human Plates were incubated at 37°C (5% CO2/95% O2) for 4 h and read by
peripheral blood was incubated with the labeled antibody at its EC50 a fluorescence reader (Molecular Devices, Sunnyvale, CA). Experi-
and the unlabeled antibody at the indicated range of concentrations ments were performed in triplicate, included isotype control antibod-
in a final volume of 200 ␮l. Plates were incubated at 4°C for 30min ies, and were repeated at least three times. Relative fluorescence
and cells were then washed. Red blood cells were lysed, washed, and units were plotted against antibody concentration and IC50 values
stained with antibodies specific for memory CD4⫹ T lymphocytes were determined from these graphs by use of GraphPad Prism Ver-
(CD4, CD45RO) and B lymphocytes (CD19). Binding to either mem- sion 4 nonlinear regression curve fits.
ory CD4⫹ T or B lymphocytes was examined by flow cytometry with Inhibition of MAdCAM-1 Binding to CD4ⴙ Memory T Lym-
use of a FACSCalibur flow cytometer and CellQuest Pro software. phocytes. Vedolizumab inhibition of high-affinity binding of MAd-
The geometric mean fluorescence intensity (GMFI) values of the CAM-1 to human peripheral blood memory CD4⫹ T lymphocytes was
positive memory CD4⫹ T-lymphocyte population or the entire B- tested. Peripheral blood (90 ␮l) was incubated with a saturating
lymphocyte population were plotted against antibody concentration. concentration (3 ␮g/ml) of MAdCAM-1-murine-Fc fusion protein and
In binding competition experiments, GMFI data were plotted as 4 mM MnCl2 in a final volume of 100 ␮l for 1 h at room temperature,
percentage inhibition versus antibody concentration. EC50 (for sat- in the presence or absence of vedolizumab. This saturating concen-
uration binding curves) or median inhibition concentration (IC50; for tration was determined from previous MAdCAM-1-Fc protein bind-
inhibition of binding curves) values were determined from these ing saturation curves with three independent donors. After washing
graphs by use of GraphPad Prism Version 4 (GraphPad Software with assay buffer (25 mM Tris, 4 mM MnCl2, 2.7 mM KCl, 150 mM
Inc., San Diego, CA) nonlinear regression curve fits. NaCl, 0.5% BSA, pH 7.2), the cells were stained with fluorescent-
Intracellular Staining of Interleukin-17. Memory T-helper 17 labeled anti-mouse IgG for 15 min at room temperature. After wash-
(Th17) cells (CD4⫹CD45RO⫹IL17A⫹) were identified from periph- ing again, cells were incubated with mouse serum for 10 min at room
eral blood of healthy donors. Peripheral blood mononuclear cells temperature, followed by staining with anti-CD4 and anti-CD45RO
(PBMCs) were isolated from heparinized human blood by standard antibodies for 15 min at room temperature. After washing, red blood
density-gradient procedures with use of Ficoll-Paque Plus (GE cells were lysed with BD FACS lysing solution and analyzed by flow
Healthcare, Little Chalfont, Buckinghamshire, UK). PBMCs were cytometry in a FACSCalibur with CellQuest Pro software. Antibody
resuspended in assay buffer for CD4 memory cell interleukin (IL) dose-dependent inhibition curves were obtained by plotting the per-
17A intracellular staining (eBioscience) according to the manufac- centage of memory CD4⫹ T lymphocytes that bound MAdCAM-1
turer’s instructions. PBMCs were stimulated (or not stimulated, for versus antibody concentration with use of GraphPad Prism Version
the control samples) with 50 ng/ml phorbol 12-myristate 13-acetate 4. IC50 values were determined from these graphs with use of Graph-
(Sigma-Aldrich) and 1 ␮g/ml Ionomycin (Sigma-Aldrich) in the pres- Pad Prism Version 4 nonlinear regression curve fits.
ence of 3 ␮M Monensin (Sigma-Aldrich) for 5 h at 37°C. After wash-
ing, cells were stained with fluorescent-labeled antibodies: vedol- Results
izumab, anti-␣4 (CD49d; 9F10) and anti-␤7 (FIB504), anti-CD4
(SK3), and anti-CD45RO (UCHL1), where applicable, at 4°C for 30 Binding of Vedolizumab to Human Tissue ex Vivo.
min. Cells were then washed with cold PBS and resuspended in The binding specificity of vedolizumab was investigated in 38
fixation and permeabilization working solution (eBioscience) stain- different types of normal human tissue by immunohisto-
Binding and Inhibitory Properties of Vedolizumab 867
chemistry. Binding of vedolizumab was restricted to the cell and markers of leukocyte subsets (including memory and
membrane of mononuclear cells in lymphoid tissues, mono- naive CD4 and CD8 T lymphocytes, B lymphocytes, natural
nuclear infiltrates in tissues of the GI tract and bladder, and killer cells, monocytes, basophils, eosinophils, and neutro-
mononuclear cells in the lumens of blood vessels (Table 1). In phils). Experiments were designed so that coexpression of the
GI tract tissues, vedolizumab bound mononuclear cells orga- ␣4␤1 and ␣4␤7 integrins and binding of vedolizumab to sub-
nized in submucosal lymphoid nodules and/or scattered in sets expressing the ␣4␤1 integrin could also be examined.
lamina propria. The large and small intestine contained the Vedolizumab bound to the majority of B lymphocytes, naive
highest frequency of mononuclear cells that were bound by CD4 and CD8 T lymphocytes, natural killer cells, and ba-
vedolizumab (data not shown). sophils at low to intermediate levels (Fig. 1, A and B). Ved-
Binding Specificity of Vedolizumab to Human Leu- olizumab bound to B lymphocytes at intermediate levels uni-
kocytes ex Vivo. Flow cytometry experiments were per- formly, to natural killer cells and basophils at low levels
formed on human peripheral blood stained with vedolizumab uniformly, and to naive CD4 and CD8 T lymphocytes from
or Act-1, anti-␣4- and anti-␤1-monoclonal antibodies (mAbs), low to intermediate levels. Vedolizumab bound to approxi-

TABLE 1
Binding specificity of vedolizumab in normal human tissues
Data are mean values from three samples from three independent donors: 0.0 to 0.3 are equivocal), 0.3 to 1.0 are weak, 1.3 to 2.0 are moderate, 2.3 to 3.0 are strong, and
3.3 to 4.0 are intense.

Vedolizumab Isotype Control


Tissue Region
20 ␮g/ml 2 ␮g/ml 20 ␮g/ml 2 ␮g/ml

Adrenal Stromal elements Neg. Neg. Neg. Neg.


Blood cells Mononuclear cells 1.0 1.0 Neg. Neg.
Polymorphonuclear cells Neg. Neg. Neg. Neg.
Platelets Neg. Neg. Neg. Neg.
Blood vessel endothelium Stromal elements Neg. Neg. Neg. Neg.
Bone marrow Stromal elements Neg. Neg. Neg. Neg.
Brain cerebrum Stromal elements Neg. Neg. Neg. Neg.
Brain cerebellum Stromal elements Neg. Neg. Neg. Neg.
Breast Stromal elements Neg. Neg. Neg. Neg.
Colon Mononuclear infiltrate 3.0 3.0 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Cervix Mononuclear infiltrate 0.5 0.5 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Esophagus Mononuclear infiltrate 2.0 1.0 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Eye Stromal elements Neg. Neg. Neg. Neg.
Heart Stromal elements Neg. Neg. Neg. Neg.
Kidney Stromal elements Neg. Neg. Neg. Neg.
Liver Stromal elements Neg. Neg. Neg. Neg.
Lung Stromal elements Neg. Neg. Neg. Neg.
Lymph node Mononuclear infiltrate 1.8 2.2 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Mammary gland Stromal elements Neg. Neg. Neg. Neg.
Ovary Stromal elements Neg. Neg. Neg. Neg.
Fallopian tube oviduct Stromal elements Neg. Neg. Neg. Neg.
Pancreas Stromal elements Neg. Neg. Neg. Neg.
Parathyroid Stromal elements Neg. Neg. Neg. Neg.
Peripheral nerve Stromal elements Neg. Neg. Neg. Neg.
Pituitary Stromal elements Neg. Neg. Neg. Neg.
Placenta Stromal elements Neg. Neg. Neg. Neg.
Prostate Mononuclear infiltrate 2.0 2.0 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Salivary gland Mononuclear infiltrate 2.2 1.7 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Skin Stromal elements Neg. Neg. Neg. Neg.
Small intestine Mononuclear infiltrate 2.7 2.7 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Spinal cord Stromal elements Neg. Neg. Neg. Neg.
Spleen Mononuclear infiltrate 2.5 2.5 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Stomach Mononuclear infiltrate 2.7 2.7 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Skeletal Muscle Stromal elements Neg. Neg. Neg. Neg.
Testis Stromal elements Neg. Neg. Neg. Neg.
Thymus Mononuclear infiltrate 2.2 2.2 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Thyroid Stromal elements Neg. Neg. Neg. Neg.
Tonsil Mononuclear infiltrate 2.2 2.2 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Urinary Bladder Mononuclear infiltrate 1.7 1.7 Neg. Neg.
Stromal elements Neg. Neg. Neg. Neg.
Uterus Stromal elements Neg. Neg. Neg. Neg.
Neg., negative.
868 Soler et al.

Fig. 1. Binding specificity of vedolizumab to human leukocytes. Human peripheral blood was stained with vedolizumab and antibodies specific for the
various leukocyte subsets and examined by flow cytometry. A, flow cytometry plots illustrating vedolizumab binding to various subsets of leukocytes,
identified by gating on the appropriate cellular markers. Vedolizumab binding is plotted against forward scatter, with the horizontal line indicating
the level of background signal as determined from a negative control antibody. Data are from at least five unrelated, healthy donors. B, the geometric
mean fluorescence intensity of vedolizumab binding to various subsets of leukocytes. Gating is restricted to the population of the leukocytes that stains
positively with vedolizumab. Data are the means from five unrelated, healthy donors with standard deviations indicated by the error bars. GMFI
values for each subset were compared with the GMFI of all leukocytes by a two-tailed, homoscedastic Student’s t test. ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍ 0.01.

mately 25% of the memory (CD45RO⫹) CD4⫹ T lymphocytes tigen specificity between these two antibodies was also dem-
at high levels (␣4␤7hi) and to 5 to 10% at intermediate levels, onstrated by the complete inhibition of labeled Act-1 binding
and not to the rest of the population (Figs. 1, A and B, and to all leukocytes exposed to unlabeled vedolizumab.
2A). Vedolizumab bound to the majority of the naive The binding profile of vedolizumab contrasts the expres-
(CD45RO⫺) CD4⫹ and CD8⫹ T lymphocytes, and to approx- sion profile of the ␣4␤1 integrin. Expression of the ␣4␤1 inte-
imately 50% of the memory (CD45RO⫹) CD8⫹ T lympho- grin, in general, was more widespread than binding by ved-
cytes, at low to intermediate levels (Fig. 1, A and B, and 2A). olizumab. Vedolizumab bound naive CD4⫹ T lymphocytes at
Vedolizumab bound to all eosinophils at intermediate to high varying levels (Fig. 2A), whereas these cells expressed the
levels, and to approximately15% of monocytes at low levels, ␣4␤1 integrin uniformly (Fig. 2B). Vedolizumab bound to a
but did not bind to neutrophils (Fig. 1, A and B). The highest specific subset of the memory CD4⫹ T-lymphocyte popula-
level of binding by vedolizumab was observed on the ␣4␤7hi tion; vedolizumab bound approximately 25% of memory
population of memory CD45RO⫹ CD4⫹ T lymphocytes (Fig. CD4⫹ T lymphocytes at high levels (Fig. 2A), whereas most of
1B). The specificity of binding to the ␣4␤7 integrin by vedol- these cells expressed relatively high levels of the ␣4 and/or ␤1
izumab was verified by competition with Act-1; unlabeled chains (Fig. 2B). Three major subsets of memory CD4⫹ T
Act-1 completely inhibited binding of vedolizumab on all lymphocytes can be defined by ␣4␤1 integrin expression: ␣4⫺,
leukocyte subsets examined (data not shown). The same an- ␣4hi␤1hi, and ␣4hi␤1lo. Vedolizumab binds to most (⬃80%) of the
Binding and Inhibitory Properties of Vedolizumab 869

Fig. 2. A comparison of the binding of


vedolizumab to expression of the ␣4 and
␤1 chains by human CD4⫹ T lympho-
cytes and monocytes. Human periph-
eral blood was stained with the indi-
cated antibodies and examined by flow
cytometry. A, blood was stained with
vedolizumab, and anti-CD4 and anti-
CD45RO antibodies. These plots illus-
trate the binding of vedolizumab in the
total (CD4⫹), naive (CD4⫹CD45RO⫺),
and memory (CD4⫹CD45RO⫹) CD4⫹
T-cell subsets. The box indicates the
level of background signal of the nega-
tive population, and the horizontal line
denotes the ␣4␤7hi population. B, blood
was stained with anti-CD4, -CD45RO,
-␣4, and -␤1 antibodies. These plots il-
lustrate the coexpression of the ␣4 and
␤1 chains in the total, naive, and mem-
ory CD4⫹ T-cell subsets. C, blood was
stained with vedolizumab, and anti-
CD4, -␣4, and -␤1 antibodies. Vedol-
izumab binding was examined in the
three ␣4⫹␤1⫹ CD4⫹ memory T-cell pop-
ulations of the total CD4⫹ T-cell plot
of B: ␣4hi␤1lo (I), ␣4hi␤1hi (II), and ␣4lo␤1hi
(III). D, blood was stained with vedol-
izumab and anti-CD14 antibodies. This
plot illustrates the binding of vedol-
izumab to monocytes. E, specificity of
binding was demonstrated by competi-
tion with Act-1. F, blood was stained
with anti-CD14, -␣4, and -␤1 antibodies.
This plot illustrates coexpression of ␣4
and ␤1 by monocytes. All data are rep-
resentative of at least five unrelated,
healthy donors.

␣4hi␤1lo T-helper lymphocytes at high levels (Fig. 2C); this is Vedolizumab Specificity for the ␣4␤7 Integrin Versus
the subset that contains the majority of the ␣4␤7hi subset. In the ␣4␤1 and ␣E␤7 Integrins. The specificity of vedol-
contrast, the majority (⬃90%) of ␣4hi␤1hi and all ␣4lo␤1hi T-helper izumab binding for the integrin ␣4␤7 versus ␣4␤1 in whole
lymphocytes are not bound by vedolizumab (Fig. 2C). Like- blood is illustrated by comparing two subsets of leukocytes
wise, a majority (⬃85%) of monocytes are not bound by that express high levels of the ␣4␤1 integrin but do not bind
vedolizumab (Fig. 2, D and E), but express relatively high vedolizumab, specifically the ␣4␤1hi subset of the memory
levels of the ␣4␤1 integrin (Fig. 2F). CD4 population (Fig. 2B, plot 1, quadrant II) and monocytes
870 Soler et al.

Fig. 3. The specificity of vedolizumab binding to the ␣4␤1, ␣4␤7, and ␣E␤7 integrins. A, flow cytometric analysis of vedolizumab binding to cell lines
that express exclusively the ␣4␤7 (A and B), ␣4␤1 (C and D), and ␣E␤7 (E and F) integrins. The flow cytometry plots represent at least three
independent experiments.

(Fig. 2F). Vedolizumab did not bind the vast majority of cells memory Th17 cell population expressed the ␣4 chain (␣4⫹␤1⫹
in either ␣4␤1hi populations (Fig. 2C, plot II, and D, respec- and ␣4⫹␤7⫹; Fig. 4). The Th17 cell population therefore con-
tively). The lack of binding by vedolizumab was maintained sists of three major subsets: ␣4⫺ (20%), ␣4␤1hi (60%), and
up to a concentration of 400 ␮g/ml (data not shown). The ␣4␤7hi (20%).
analysis of the specificity of vedolizumab for the ␣4␤7 integrin Potency of Binding of Vedolizumab to B and Memory
versus ␣E␤7 in whole blood by flow cytometry was not possi- CD4ⴙ T Lymphocytes. The potency of vedolizumab for
ble because all the ␣E␤7-expressing cells are contained within binding to human peripheral blood B and memory CD4⫹
the ␣4␤7hi subset. The specificity of binding to integrins con- lymphocytes was estimated from saturation binding curves
taining the ␣4 and/or ␤7 chains by vedolizumab was investi- in experiments with labeled vedolizumab (Fig. 5A). The EC50
gated further with cell lines expressing the ␣4␤1, ␣4␤7, or values were 0.067 ␮g/ml (0.4 nM) and 0.042 ␮g/ml (0.3 nM)
␣E␤7 integrins exclusively. Vedolizumab bound to RPMI8866 for B and memory CD4⫹ lymphocytes, respectively. The po-
cells (Fig. 3A) that expressed the ␣4 and ␤7 chains (Fig. 3B) tency of vedolizumab for binding to human peripheral blood
but not the ␤1 or ␣E chains (not shown). Incubation with B and memory CD4⫹ lymphocytes was also estimated from
Act-1 competed with the binding of vedolizumab to competitive binding experiments in which binding of labeled
RPMI8866 cells, confirming the specificity of binding to the vedolizumab at its EC50 was competed off by unlabeled ved-
␣4␤7 integrin (data not shown). Vedolizumab did not bind to olizumab (Fig. 5B). The mean IC50 values were 0.045 ␮g/ml
RAMOS cells (Fig. 3C) that expressed the ␣4 and ␤1 chains (0.3 nM) and 0.044 ␮g/ml (0.3 nM) for B and memory CD4⫹
(Fig. 3D) but not the ␤7 chain (data not shown), nor to mouse lymphocytes, respectively. Similar results were obtained
L1.2 cell transfectants (Fig. 3E) that expressed the human ␣E with Act-1; the IC50 values for Act-1 binding were 0.062
and ␤7 chains (Fig. 3, E and F), but not the human ␣4 chain ␮g/ml (0.010 nM) and 0.059 ␮g/ml (0.008 nM) for B and
(data not shown). memory CD4⫹ lymphocytes, respectively (data not shown).
Characterization of the Memory CD4 T Lymphocyte Vedolizumab Inhibits Adhesion of ␣4␤7ⴙ Cells to
Population for Expression of the ␣4␤1 and ␣4␤7 Inte- MAdCAM-1 and Fibronectin but Not to VCAM-1. The
grins and IL-17. A subset (⬃20%) of the Th17 cell popula- neutralizing potency and specificity of vedolizumab was de-
tion in peripheral blood was bound by vedolizumab (Fig. 4), termined in cell adhesion assays with ␣4␤7-expressing
and similar data were obtained for expression of the ␤7 chain RPMI8866 cells and the cell adhesion proteins, MAdCAM-1,
(data not shown). In contrast, the majority (80%) of the VCAM-1, and the alternatively spliced form of fibronectin
Binding and Inhibitory Properties of Vedolizumab 871

100 0.058 ⫾ 0.024 ␮g/ml (0.39 ⫾ 0.16 nM; n ⫽ 6). Vedolizumab


inhibited low-affinity adhesion of ␣4␤7-expressing RPMI8866

% Of Total Th17 Population


80 cells (no Mn2⫹ activation) to MAdCAM-1 with similar
potency (0.023 ⫾ 0.012 ␮g/ml; 0.15 ⫾ 0.08 nM; n ⫽ 7; Fig.
60
6B). Vedolizumab inhibited adhesion of ␣4␤7-expressing
RPMI8866 cells to fibronectin (Mn2⫹-activated, as required
for fibronectin binding; Fig. 6F) with a mean IC50 value of
40
0.02 ⫾ 0.012 ␮g/ml (0.14 ⫾ 0.08 nM; n ⫽ 4). In contrast,
** ** vedolizumab did not inhibit adhesion of ␣4␤7-expressing
20
RPMI8866 cells to VCAM-1 under high- (Fig. 6, C, n ⫽ 4, and
E) or low-affinity (Fig. 6, D, n ⫽ 3, and E) states, even at 400
0 ␮g/ml, the highest concentration tested (Fig. 6E, n ⫽ 3). The

α4-
α4+
vedolizumab-
vedolizumab+

anti-␣4-mAb, in contrast, inhibited all adhesion of ␣4␤7-ex-


pressing RPMI8866 cells with MAdCAM-1, VCAM-1, and
fibronectin with subnanomolar potency (Fig. 6, A–F). The
humanized mAb negative control (Fig. 6, A, B, and F) and the
murine isotype-matched negative control (Fig. 6, A–F) had no
Fig. 4. Vedolizumab binds to a subset of Th17 lymphocytes. Flow cytometric effect on adhesion. The effect of vedolizumab on the binding
analysis illustrates the percentage of human peripheral blood Th17 lympho- of MAdCAM-1 to T lymphocytes in human whole blood was
cytes that bind vedolizumab and anti-␣4-antibody. Data are the means of at also evaluated by flow cytometry. Vedolizumab inhibited the
least four unrelated, healthy donors with standard deviations indicated by
the error bars. Values for percentage of total Th17 population for the positive binding of soluble human MAdCAM-1-Fc fusion protein to
(⫹) population were compared with the negative (⫺) population by use of a the ␣4␤7hi CD4 memory cell population with similar potency
two-tailed, homoscedastic Student’s t test. ⴱ, p ⬍ 0.05; ⴱⴱ, p ⬍ 0.01. (IC50 ⫽ 0.034 ␮g/ml, 0.225 nM mean of three donors) (Fig. 7).
Vedolizumab Has No Effect on Adhesion of the ␣4␤1
A 80 Integrin to VCAM-1 or Fibronectin. The specificity of
70 vedolizumab was further characterized in adhesion assays
with the ␣4␤1-expressing RAMOS cells. Vedolizumab did not
60
inhibit high- (Mn2⫹-activated) or low-affinity (no Mn2⫹) ad-
50 hesion of ␣4␤1-expressing RAMOS cells to VCAM-1 (Fig. 8, A
GMFI

40 and C, respectively), even at the highest concentrations as-


sayed, 400 ␮g/ml (Fig. 8, B and D, respectively). Conversely,
30
anti-␣4-mAb, anti-␤1-mAb, and a combination thereof, inhib-
20 ited all adhesion between ␣4␤1-expressing RAMOS cells and
10 VCAM-1 (Fig. 8, A–D). Vedolizumab did not inhibit adhesion
of ␣4␤1-expressing RAMOS cells to fibronectin at 400 ␮g/ml,
0
the highest concentration assayed, whereas a combination of
-10 -3 -2 -1 0 1 2
10 10 10 10 10 10 anti-␣4- and anti-␤1-mAbs inhibited adhesion (Fig. 8E).
Vedolizumab (µµg/ml) RAMOS cells did not bind to MAdCAM-1, even after integrin
B 120 activation with Mn2⫹ (data not shown).

100

80 Discussion
% Inhibition

60 Infiltration of the GI tract by T lymphocytes is a patho-


genic mechanism of UC and CD (Xavier and Podolsky, 2007).
40 Migration into the GI tract is a complex, multistep process
requiring the coordinated interaction of several adhesion and
20
signaling molecules (selectins, integrins, chemokine recep-
0 tors) on the surface of T lymphocytes, with their correspond-
ing ligands on the endothelium (Salmi and Jalkanen, 2005).
10
-5
10
-4
10
-3
10
-2
10
-1
10
0
10
1
10
2 The ␣4␤7 integrin is a pivotal mediator of infiltration of GI
tract by T lymphocytes and antagonizing its adhesion to
µg/ml)
Vedolizumab (µ
MAdCAM-1 provides efficacy in animal models of IBD (Hes-
Fig. 5. Binding of vedolizumab to human peripheral blood B and memory
CD4⫹ T lymphocytes. A, saturation binding curve of vedoliumab-alexa-647
terberg et al., 1996; Picarella et al., 1997) and in patients
binding to peripheral blood B lymphocytes (F) and memory CD4⫹ T lym- with UC (Feagan et al., 2005, 2008) and CD (Feagan et al.,
phocytes (E). B, dose-dependent inhibition curve of vedolizumab-alexa-647 2005, 2008). These investigations also demonstrated that
binding to B lymphocytes (F) and memory CD4⫹ lymphocytes (E) by unla- antagonizing the ␣4␤7 integrin is well tolerated. The absence
beled vedolizumab. Control antibody for B lymphocytes (f) and memory
CD4⫹ T lymphocytes (䡺). The data represent multiple donors. of overt deleterious effects is partially attributable to vedol-
izumab binding exclusively to leukocytes. This binding pro-
containing the CS-1 peptide. Vedolizumab inhibited high- file is consistent with the expression profile of the human
affinity adhesion of ␣4␤7-espressing RPMI8866 cells (Mn2⫹- ␣4␤7 integrin (Schweighoffer et al., 1993; Erle et al., 1994;
activated) to MAdCAM-1 (Fig. 6A) with a mean IC50 value of Farstad et al., 1997; Rott et al., 2000).
872 Soler et al.

Fig. 6. Potency and specificity of vedolizumab


antagonism of ␣4␤7 integrin adhesion to MAd-
CAM-1 and fibronectin, but not to VCAM-1. Ef-
fect of vedolizumab (F, A–D, F), a humanized
mAb negative control (‚, A–B, F), an anti-␣4-
mAb (F, A–D, F), and isotype control Ig (aster-
isk, A–D, F) on adhesion of ␣4␤7-expressing
RPMI8866 cells to MAdCAM-1 with high (A) and
low (B) affinity, to VCAM-1 with high (C and E)
and low (D and E) affinity, and to fibronectin
with high affinity (F). In E, data are the mean of
three independent experiments with standard
deviations indicated by the error bars. Relative
fluorescent unit (RFU) values for cells incubated
with antibody were compared with RFU values
for cells not incubated with antibody with use of
a two-tailed, homoscedastic Student’s t test. ⴱ,
p ⬍ 0.05; ⴱⴱ, p ⬍ 0.01.

ducing inflammation in diseased tissue. The second strategy


MAdCAM-1+-CD4+ Memory cells (%)

30
targets the ␣4 chain and inhibits both the ␣4␤7 and ␣4␤1
integrins. The ␣4␤1 integrin is expressed by all leukocytes
except neutrophils (González-Amaro et al., 2005), and antag-
20
onism by natalizumab inhibits adhesion of the ␣4␤1 integrin
to VCAM-1, fibronectin, and osteopontin, and the adhesion of
the ␣4␤7 integrin to MAdCAM-1, VCAM-1, and fibronectin
10 (Biogen Idec, 2006). The ␣4␤1 integrin mediates adhesion in
many different types of tissue, and inhibition by natalizumab
therefore induces diverse systemic effects, such as mobiliza-
0 tion of hematopoietic progenitor cells from the bone marrow
10 -1 10 0 10 1 10 2 10 3 10 4 (Bonig et al., 2008; Zohren et al., 2008), leukocytosis in the
µg/ml)
Vedolizumab (µ vasculature (Ghosh et al., 2003; Sandborn et al., 2005; Tar-
Fig. 7. Vedolizumab antagonism of MAdCAM-1-Fc binding to the ␣4␤7hi gan et al., 2007), and reduction in the number of leukocytes
CD4 memory T-lymphocyte population in human peripheral blood in in cerebral spinal fluid (Stüve et al., 2006a,b) and cerebral
vitro. Blood was stained with MAdCAM-1-Fc, with or without vedol-
izumab, and with anti-CD4 and anti-CD45RO antibodies. MAdCAM-1-Fc tissue (del Pilar Martin et al., 2008). This diverse profile of
binding to the CD4 memory T-lymphocyte population was analyzed by physiologic effects may confer efficacy in pathologically dis-
flow cytometry. Results are reported as the percentage of the total CD4 tinct diseases, such as multiple sclerosis (Miller et al., 2003)
memory T-lymphocyte population that bound MAdCAM-1 versus vedol-
izumab concentration. Data are the mean of three independent donors. and CD (Ghosh et al., 2003; Sandborn et al., 2005; Targan et
al., 2007). Repeated administration of natalizumab to pa-
This desirable profile encouraged targeting the ␣4␤7 inte- tients with multiple sclerosis and CD, however, is also asso-
grin by three different therapeutic strategies. One of these ciated with systemic immunosuppression, including an in-
strategies targets the ␤7 chain of the integrin and, conse- creased incidence of the fatal infectious disease, PML
quently, inhibits both the ␣4␤7 and ␣E␤7 adhesion pathways (Kleinschmidt-DeMasters and Tyler, 2005; Langer-Gould et
thereby inhibiting the localization and retention of T lympho- al., 2005; Van Assche et al., 2005). It is postulated that the
cytes within epithelial layers in numerous tissues (Kilshaw, anti-inflammatory mechanism that mediates efficacy of na-
1999). This could potentially elicit systemic effects, perturb- talizumab in multiple sclerosis and CD may also predispose
ing immunosurveillance of normal epithelial tissue and re- patients to PML by decreasing immunosurveillance in the
Binding and Inhibitory Properties of Vedolizumab 873

Fig. 8. Effect of vedolizumab (F, A and C), anti-


␣4-mAb (E, A and C), and isotype control Ig
(asterisk, A and C) on adhesion of cells express-
ing the ␣4␤1 integrin to VCAM-1 and fibronectin.
Effect of mAbs on high- (A and B) and low-
affinity (C and D) binding of ␣4␤1-expressing
RAMOS cells to VCAM-1, and effect of mAbs on
binding to fibronectin (E). Anti-␣4-mAb, anti-␤1-
mAb, mouse IgG at 10 ␮g/ml, and vedolizumab
at 400 ␮g/ml were used (B, D, and E). In B, D,
and E, data are the mean of three independent
experiments with standard deviations indicated
by the error bars. RFU values for cells incubated
with antibody were compared with RFU values
for cells incubated with mouse IgG using a two-
tailed, homoscedastic Student’s t test. ⴱ, p ⬍
0.05; ⴱⴱ, p ⬍ 0.01.

central nervous system (Berger and Houff, 2006; Stüve et al., Differences in expression of integrin chains between mem-
2006a,b; del Pilar Martin et al., 2008). Natalizumab may ory CD4⫹ T lymphocytes also contribute to the unique clini-
prevent the entry of memory T lymphocytes into the brain cal profile of vedolizumab. Circulating effector memory CD4⫹
and perhaps into sites of viral latency, thereby precluding T T lymphocytes can be divided into three subpopulations ac-
lymphocytes from clearing or suppressing the viral infection cording to expression of the ␣4 and ␤1 chains: 1) 20% that do
(Berger, 2006; Stüve et al., 2006b; del Pilar Martin et al., not express the ␣4 chain (Fig. 2B); 2) 50% that express high
2008). levels of ␣4 and ␤1 chains (Fig. 2B) that would be bound by an
Vedolizumab uses a third strategy: exclusive targeting of anti-␣4-therapeutic but not by vedolizumab (Fig. 2C), and
the ␣4␤7 integrin. Vedolizumab binds to the ␣4␤7 integrin, 3) 30% that express high levels of the ␣4␤7 integrin and low
but not to the ␣4␤1 or ␣E␤7 integrins (Figs. 2 and 3). This levels of the ␤1 chain (Fig. 2C) that would be bound by both
specificity was confirmed in functional assays demonstrating an anti-␣4-therapeutic and vedolizumab. The CD4⫹ memory
that vedolizumab inhibits adhesion of ␣4␤7-expressing cells ␣4␤7hi population is postulated to be pathogenic in IBD
exclusively (Figs. 6 and 7), but not of ␣4␤1-expressing cells (Butcher et al., 1999; Salmi and Jalkanen, 2005), and the
exclusively (Fig. 8). These data agree with immunoprecipita- clinical efficacy of natalizumab in CD and vedolizumab in CD
tion experiments that utilized Act-1 (Schweighoffer et al., and UC buttress this paradigm (Ghosh et al., 2003; Feagan et
1993). Specifically targeting the ␣4␤7 integrin enables vedol- al., 2005, 2008; Sandborn et al., 2005). Vedolizumab specifi-
izumab to immunomodulate the GI tract without systemic cally targets the CD4⫹ memory subpopulation that is patho-
effects, due to the GI-specific role of the ␣4␤7 integrin in genic in IBD (␣4␤7hi), while sparing other CD4⫹ memory
mediating infiltration by leukocytes (Butcher et al., 1999; subpopulations (␣4␤1hi) and monocytes (Fig. 2) that are inte-
Engelhardt and Briskin, 2005; Agace, 2006). This strategy gral for immunosurveillance and host defense.
does not elicit leukocytosis in UC and CD patients, and has We discovered that vedolizumab binds to a subset (⬃20%)
demonstrated an excellent clinical safety profile to date of Th17 cells that express the gut-tropic ␣4␤7hi phenotype
(Feagan et al., 2005, 2008). (Fig. 4). This proinflammatory subset is found in numerous
Another component of the unique pharmacologic activity of types of inflamed tissue and is postulated to mediate many
vedolizumab could be selective inhibition of cellular activity. different autoimmune diseases, such as psoriasis, psoriatic
Vedolizumab selectively inhibits the adhesion of cells ex- arthritis, rheumatoid arthritis, type 1 diabetes, transplant
pressing the ␣4␤7 integrin to MAdCAM-1 and fibronectin, but rejection, and tumor immunotherapy (Iwakura and Ishig-
not to VCAM-1 (Figs. 6 and 7). These data are consistent with ame, 2006). IL-23 plays an important role in the maintenance
qualitative reports for the effect of Act-1 on adhesion of B and and function of the Th17 subset of CD4 memory T lympho-
T lymphocytes to these ligands (Postigo et al., 1993; Schweig- cytes. Single-nucleotide polymorphisms of the human IL-23
hoffer et al., 1993; Erle et al., 1994; Walsh et al., 1996). receptor gene are associated with increased risk and protec-
874 Soler et al.

tion from CD (Duerr et al., 2006). Ustekinumab is a thera- Farstad IN, Halstensen TS, Kvale D, Fausa O, and Brandtzaeg P (1997) Topographic
distribution of homing receptors on B and T cells in human gut-associated lym-
peutic antibody targeting the human p40 subunit of the IL-12 phoid tissue: relation of L-selectin and integrin alpha 4 beta 7 to naive and memory
and IL-23 cytokines, and is efficacious in CD (Sandborn et al., phenotypes. Am J Pathol 150:187–199.
Feagan BG, Greenberg GR, Wild G, Fedorak RN, Paré P, McDonald JW, Cohen A,
2008) and psoriasis (Leonardi et al., 2008; Papp et al., 2008). Bitton A, Baker J, Dubé R, et al. (2008) Treatment of active Crohn’s disease with
It seems that vedolizumab targets only a subset (20%) of the MLN0002, a humanized antibody to the alpha4beta7 integrin. Clin Gastroenterol
Th17 cells, namely the GI-tropic, ␣4␤7hi subset and thus could Hepatol 6:1370 –1377.
Feagan BG, Greenberg GR, Wild G, Fedorak RN, Paré P, McDonald JW, Dubé R,
provide efficacy in IBD more specifically than ustekinumab, Cohen A, Steinhart AH, Landau S, et al. (2005) Treatment of ulcerative colitis with
conferring an improved clinical risk-to-benefit profile in pa- a humanized antibody to the alpha4beta7 integrin. N Engl J Med 352:2499 –2507.
Ghosh S, Goldin E, Gordon FH, Malchow HA, Rask-Madsen J, Rutgeerts P, Vyh-
tients with IBD. nálek P, Zádorová Z, Palmer T, and Donoghue S (2003) Natalizumab for active
Targeting systemic mediators of inflammation, such as Crohn’s disease. N Engl J Med 348:24 –32.
González-Amaro R, Mittelbrunn M, and Sánchez-Madrid F (2005) Therapeutic anti-
␣4hi␤1hi T cells and Th17 cells, inhibits systemic immunosur- integrin (alpha4 and alphaL) monoclonal antibodies: two-edged swords? Immunol-
veillance, which may predispose patients to infection and/or ogy 116:289 –296.
Hesterberg PE, Winsor-Hines D, Briskin MJ, Soler-Ferran D, Merrill C, Mackay CR,
neoplasia (Engelhardt and Briskin, 2005; Ransohoff, 2005; Newman W, and Ringler DJ (1996) Rapid resolution of chronic colitis in the
Iwakura and Ishigame, 2006; Wei et al., 2006; Mottet and cotton-top tamarin with an antibody to a gut-homing integrin alpha 4 beta 7.
Golshayan, 2007). The role of the ␣4␤7 integrin in immuno- Gastroenterology 111:1373–1380.
Iwakura Y and Ishigame H (2006) The IL-23/IL-17 axis in inflammation. J Clin
surveillance, in contrast, is restricted to the GI tract (Butcher Invest 116:1218 –1222.
et al., 1999; Salmi and Jalkanen, 2005; Agace, 2006). Gut- Kilshaw PJ (1999) Alpha E beta 7. Mol Pathol 52:203–207.
Kleinschmidt-DeMasters BK and Tyler KL (2005) Progressive multifocal leukoen-
specific immunomodulation by vedolizumab is hence less cephalopathy complicating treatment with natalizumab and interferon beta-1a for
likely to predispose patients to infection and/or neoplasia multiple sclerosis. N Engl J Med 353:369 –374.
Koralnik IJ (2006) Progressive multifocal leukoencephalopathy revisited: has the
outside of the GI tract than anti-␣4, anti-␤7, and anti-p40 disease outgrown its name? Ann Neurol 60:162–173.
(IL-12/IL-23) subunit therapeutics. The specificity of vedol- Langer-Gould A, Atlas SW, Green AJ, Bollen AW, and Pelletier D (2005) Progressive
izumab for GI-tropic T lymphocytes offers clinical efficacy in multifocal leukoencephalopathy in a patient treated with natalizumab. N Engl
J Med 353:375–381.
IBD (Feagan et al., 2005, 2008) without some of the undesir- Lazarovits AI, Moscicki RA, Kurnick JT, Camerini D, Bhan AK, Baird LG, Erikson
able systemic effects characteristic of natalizumab and M, and Colvin RB (1984) Lymphocyte activation antigens. I. A monoclonal anti-
body, anti-Act I, defines a new late lymphocyte activation antigen. J Immunol
ustekinumab. The ability to modulate inflammation specifi- 133:1857–1862.
cally within the GI tract, without systemic immunosuppres- Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y, Li S, Dooley LT,
and Gordon KB (2008) Efficacy and safety of ustekinumab, a human interleukin-
sion, is likely to confer efficacy in IBD with an improved 12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a
safety profile and risk-to-benefit ratio. randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet 371:
1665–1674.
Miller DH, Khan OA, Sheremata WA, Blumhardt LD, Rice GP, Libonati MA,
Willmer-Hulme AJ, Dalton CM, Miszkiel KA, and O’Connor PW (2003) A con-
Acknowledgments trolled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 348:
15–23.
We thank Drs. Irving Fox, Asit Parikh, and Carl Alden, Veit
Mottet C and Golshayan D (2007) CD4⫹CD25⫹Foxp3⫹ regulatory T cells: from
Schmelmer and Dr. Jennifer Elliot, Karen Repetny, Brad Nohe, Dr. basic research to potential therapeutic use. Swiss Med Wkly 137:625– 634.
Charles Baum, Gretchen Bodun, Betsy Pilmer, Yasushi Shimizu, Niino M, Bodner C, Simard ML, Alatab S, Gano D, Kim HJ, Trigueiro M, Racicot D,
Teruhisa Tanaka, Kenichiro Nogami, Ohta Sunao, Shaila Basa- Guérette C, Antel JP, et al. (2006) Natalizumab effects on immune cell responses
in multiple sclerosis. Ann Neurol 59:748 –754.
vappa, and Yasuko Tokifuji for insightful discussions and review of Papp KA, Langley RG, Lebwohl M, Krueger GG, Szapary P, Yeilding N, Guzzo C,
the manuscript. Hsu MC, Wang Y, Li S, et al. (2008) Efficacy and safety of ustekinumab, a human
interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results
from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet
References 371:1675–1684.
Picarella D, Hurlbut P, Rottman J, Shi X, Butcher E, and Ringler DJ (1997) Mono-
Agace WW (2006) Tissue-tropic effector T cells: generation and targeting opportuni- clonal antibodies specific for beta 7 integrin and mucosal addressin cell adhesion
ties. Nature Rev Immunol 6:682– 692.
molecule-1 (MAdCAM-1) reduce inflammation in the colon of scid mice reconsti-
Berger JR (2006) Natalizumab and progressive multifocal leucoencephalopathy. Ann
tuted with CD45RBhigh CD4⫹ T cells. J Immunol 158:2099 –2106.
Rheum Dis 65 (Suppl 3):iii48 –53.
Postigo AA, Sánchez-Mateos P, Lazarovits AI, Sánchez-Madrid F, and de Landázuri
Berger JR and Houff S (2006) Progressive multifocal leukoencephalopathy: lessons
MO (1993) Alpha 4 beta 7 integrin mediates B cell binding to fibronectin and
from AIDS and natalizumab. Neurol Res 28:299 –305.
vascular cell adhesion molecule-1. Expression and function of alpha 4 integrins on
Berger JR and Koralnik IJ (2005) Progressive multifocal leukoencephalopathy and
human B lymphocytes. J Immunol 151:2471–2483.
natalizumab— unforeseen consequences. N Engl J Med 353:414 – 416.
Ransohoff RM (2005) Natalizumab and PML. Nat Neurosci 8:1275.
Biogen Idec I (2006) TYSABRI® (natalizumab), Biogen Idec Inc., Cambridge, MA.
Rott LS, Briskin MJ, and Butcher EC (2000) Expression of alpha4beta7 and E-
Bonig H, Wundes A, Chang KH, Lucas S, and Papayannopoulou T (2008) Increased
numbers of circulating hematopoietic stem/progenitor cells are chronically main- selectin ligand by circulating memory B cells: implications for targeted trafficking
tained in patients treated with the CD49d blocking antibody natalizumab. Blood to mucosal and systemic sites. J Leukoc Biol 68:807– 814.
111:3439 –3441. Salmi M and Jalkanen S (2005) Lymphocyte homing to the gut: attraction, adhesion,
Butcher EC and Picker LJ (1996) Lymphocyte homing and homeostasis. Science and commitment. Immunol Rev 206:100 –113.
272:60 – 66. Sandborn WJ, Colombel JF, Enns R, Feagan BG, Hanauer SB, Lawrance IC, Pan-
Butcher EC, Williams M, Youngman K, Rott L, and Briskin M (1999) Lymphocyte accione R, Sanders M, Schreiber S, Targan S, et al. (2005) Natalizumab induction
trafficking and regional immunity. Adv Immunol 72:209 –253. and maintenance therapy for Crohn’s disease. N Engl J Med 353:1912–1925.
del Pilar Martin M, Cravens PD, Winger R, Frohman EM, Racke MK, Eagar TN, Sandborn WJ, Feagan BG, Fedorak RN, Scherl E, Fleisher MR, Katz S, Johanns J,
Zamvil SS, Weber MS, Hemmer B, Karandikar NJ, et al. (2008) Decrease in the Blank M, and Rutgeerts P (2008) A randomized trial of ustekinumab, a human
numbers of dendritic cells and CD4⫹ T cells in cerebral perivascular spaces due to interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe
natalizumab. Arch Neurol 65:1596 –1603. Crohn’s disease. Gastroenterology 135:1130 –1141.
Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, Daly MJ, Steinhart AH, Schweighoffer T, Tanaka Y, Tidswell M, Erle DJ, Horgan KJ, Luce GE, Lazarovits
Abraham C, Regueiro M, Griffiths A, et al. (2006) A genome-wide association study AI, Buck D, and Shaw S (1993) Selective expression of integrin alpha 4 beta 7 on
identifies IL23R as an inflammatory bowel disease gene. Science 314:1461–1463. a subset of human CD4⫹ memory T cells with hallmarks of gut-trophism. J Im-
Engelhardt B and Briskin MJ (2005) Therapeutic targeting of alpha 4-integrins in munol 151:717–729.
chronic inflammatory diseases: tipping the scales of risk towards benefit? Eur Stüve O, Marra CM, Bar-Or A, Niino M, Cravens PD, Cepok S, Frohman EM,
J Immunol 35:2268 –2273. Phillips JT, Arendt G, Jerome KR, et al. (2006a) Altered CD4⫹/CD8⫹ T-cell ratios
Engelhardt B, Laschinger M, Schulz M, Samulowitz U, Vestweber D, and Hoch G in cerebrospinal fluid of natalizumab-treated patients with multiple sclerosis.
(1998) The development of experimental autoimmune encephalomyelitis in the Arch Neurol 63:1383–1387.
mouse requires alpha4-integrin but not alpha4beta7-integrin. J Clin Invest 102: StüveO, Marra CM, Jerome KR, Cook L, Cravens PD, Cepok S, Frohman EM,
2096 –2105. Phillips JT, Arendt G, Hemmer B, et al. (2006b) Immune surveillance in multiple
Erle DJ, Briskin MJ, Butcher EC, Garcia-Pardo A, Lazarovits AI, and Tidswell M sclerosis patients treated with natalizumab. Ann Neurol 59:743–747.
(1994) Expression and function of the MAdCAM-1 receptor, integrin alpha 4 beta Targan SR, Feagan BG, Fedorak RN, Lashner BA, Panaccione R, Present DH,
7, on human leukocytes. J Immunol 153:517–528. Spehlmann ME, Rutgeerts PJ, Tulassay Z, Volfova M, et al. (2007) Natalizumab
Binding and Inhibitory Properties of Vedolizumab 875
for the treatment of active Crohn’s disease: results of the ENCORE Trial. Gastro- Wei S, Kryczek I, and Zou W (2006) Regulatory T-cell compartmentalization and
enterology 132:1672–1683. trafficking. Blood 108:426 – 431.
Tidswell M, Pachynski R, Wu SW, Qiu SQ, Dunham E, Cochran N, Briskin MJ, Xavier RJ and Podolsky DK (2007) Unravelling the pathogenesis of inflammatory
Kilshaw PJ, Lazarovits AI, Andrew DP, et al. (1997) Structure-function analysis of bowel disease. Nature 448:427– 434.
the integrin beta 7 subunit: identification of domains involved in adhesion to Zohren F, Toutzaris D, Klärner V, Hartung HP, Kieseier B, and Haas R (2008) The
MAdCAM-1. J Immunol 159:1497–1505. monoclonal anti-VLA-4 antibody natalizumab mobilizes CD34⫹ hematopoietic
Van Assche G, Van Ranst M, Sciot R, Dubois B, Vermeire S, Noman M, Verbeeck J, progenitor cells in humans. Blood 111:3893–3895.
Geboes K, Robberecht W, and Rutgeerts P (2005) Progressive multifocal leukoen-
cephalopathy after natalizumab therapy for Crohn’s disease. N Engl J Med 353:
362–368. Address correspondence to: Dr. Eric R. Fedyk, Millennium Pharmaceu-
Walsh GM, Symon FA, Lazarovils AL, and Wardlaw AJ (1996) Integrin alpha 4 beta ticals Inc., 40 Landsdowne Street, Cambridge, MA 02139. E-mail: fedyk@
7 mediates human eosinophil interaction with MAdCAM-1, VCAM-1 and fibronec- mpi.com
tin. Immunology 89:112–119.

Anda mungkin juga menyukai