ArticlePDF Available

Selective blockade of CD28 and not CTLA-4 with a single-chain Fv- 1-antitrypsin fusion antibody

Authors:

Abstract and Figures

B7-1 and B7-2 are costimulatory molecules expressed on antigen-presenting cells. The CD28/B7 costimulation pathway is critical for T-cell activation, proliferation, and Th polarization. Blocking both cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and CD28 interactions with a CTLA-4/Ig fusion protein inhibits various immune-mediated processes in vivo, such as allograft rejection and autoimmunity. However, selective blockade of CD28 may represent a better strategy for immunosuppression than B7 blockade, because CTLA-4/B7 interactions have been shown to participate in the extinction of the T-cell receptor-mediated activation signal and to be required for the induction of immunologic tolerance. In addition, selective CD28 inhibition specifically decreases the activation of alloreactive and autoreactive T cells, but not the activation of T cells stimulated by exogenous antigens presented in the context of self major histocompatibility complex (MHC) molecules. CD28 blockade cannot be obtained with anti-CD28 dimeric antibodies, which cluster their target and promote T-cell costimulation, whereas monovalent Fab fragments can block CD28 and reduce alloreactivity. In this study, we report the construction of a monovalent single-chain Fv antibody fragment from a high-affinity antihuman CD28 antibody (CD28.3) that blocked adhesion of T cells to cells expressing the CD28 receptor CD80. Genetic fusion with the long-lived serum protein alpha1-antitrypsin led to an extended half-life without altering its binding characteristics. The anti-CD28 fusion molecule showed biologic activity as an immuno-suppressant by inhibiting T-cell activation and proliferation in a mixed lymphocyte reaction.
Content may be subject to copyright.
doi:10.1182/blood-2002-08-2480
Prepublished online March 20, 2003;
2003 102: 564-570
Haspot, Jérôme Tiollier and Jean-Paul Soulillou
Bernard Vanhove, Geneviève Laflamme, Flora Coulon, Marie Mougin, Patricia Vusio, Fabienne
1-antitrypsin fusion antibody αSelective blockade of CD28 and not CTLA-4 with a single-chain Fv
http://bloodjournal.hematologylibrary.org/content/102/2/564.full.html
Updated information and services can be found at:
(577 articles)Immunotherapy (5022 articles)Immunobiology (790 articles)Cell Adhesion and Motility Articles on similar topics can be found in the following Blood collections
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#repub_requests
Information about reproducing this article in parts or in its entirety may be found online at:
http://bloodjournal.hematologylibrary.org/site/misc/rights.xhtml#reprints
Information about ordering reprints may be found online at:
http://bloodjournal.hematologylibrary.org/site/subscriptions/index.xhtml
Information about subscriptions and ASH membership may be found online at:
Copyright 2011 by The American Society of Hematology; all rights reserved.
Washington DC 20036.
by the American Society of Hematology, 2021 L St, NW, Suite 900,
Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly
For personal use only. by guest on June 12, 2013. bloodjournal.hematologylibrary.orgFrom
IMMUNOBIOLOGY
Selective blockade of CD28 and not CTLA-4 with a single-chain Fv–
1-antitrypsin fusion antibody
Bernard Vanhove, Genevie`ve Laflamme, Flora Coulon, Marie Mougin, Patricia Vusio, Fabienne Haspot, Je´roˆme Tiollier, and Jean-Paul Soulillou
B7-1 and B7-2 are costimulatory molecules
expressed on antigen-presenting cells. The
CD28/B7 costimulation pathway is critical
for T-cell activation, proliferation, and Th
polarization. Blocking both cytotoxic T-
lymphocyte–associated antigen 4 (CTLA-4)
and CD28 interactions with a CTLA-4/Ig
fusion protein inhibits various immune-
mediated processes in vivo, such as allo-
graft rejection and autoimmunity. However,
selective blockade of CD28 may represent a
better strategy for immunosuppression than
B7 blockade, because CTLA-4/B7 interac-
tions have been shown to participate in the
extinction of the T-cell receptor–mediated
activation signal and to be required for the
induction of immunologic tolerance. In addi-
tion, selective CD28 inhibition specifically
decreases the activation of alloreactive and
autoreactive T cells, but not the activation of
T cells stimulated by exogenous antigens
presented in the context of self major histo-
compatibility complex (MHC) molecules.
CD28 blockade cannot be obtained with
anti-CD28 dimeric antibodies, which cluster
their target and promote T-cell costimula-
tion, whereas monovalent Fab fragments
can block CD28 and reduce alloreactivity. In
this study, we report the construction of a
monovalent single-chain Fv antibody frag-
ment from a high-affinity antihuman CD28
antibody (CD28.3) that blocked adhesion of
T cells to cells expressing the CD28 recep-
tor CD80. Genetic fusion with the long-lived
serum protein 1-antitrypsin led to an ex-
tended half-life without altering its binding
characteristics. The anti-CD28 fusion mole-
cule showed biologic activity as an immuno-
suppressant by inhibiting T-cell activation
and proliferation in a mixed lymphocyte
reaction. (Blood. 2003;102:564-570)
©2003 by The American Society of Hematology
Introduction
Targeting T-cell costimulation has been widely investigated to control
T-cell reactivity in autoimmunity and transplantation. Inhibiting costimu-
lation through B7 (B7-1/B7-2 or CD80/86) blockade by cytotoxic
T-lymphocyte–associated antigen 4 (CTLA-4)–Ig has now become a
clinical reality.1Blocking B7, however, might have 2 opposite effects. In
addition to reducing T-cell costimulation through CD28, B7 blockade
can also prevent CTLA-4 from transmitting a negative/regulatory
signal. Indeed, the signal transmitted through CTLA-4 leads to the
recruitment of phosphatases that dephosphorylate activated second
messengers in the CD3 complex resulting in a reduction in the nuclear
translocation of Rel A needed for the production of multiple cytokines
produced by Th1 and Th2 cells.2,3 Moreover, CTLA-4 has been
implicated in the development of regulatory T cells in several models of
transplantation4and animals lacking CTLA-4 are resistant to tolerance
induction.5Therefore, inactivation of CTLA-4 might well be detrimen-
tal to the development of transplantation tolerance and may ultimately
oppose the effect of B7 blockade. This double-edged nature of B7
blockade suggests that an agent that would selectively block CD28
and not CTLA-4 may be more adapted to maneuvers aimed at
inducing tolerance.
Despite the clinical promise of anti-CD28–targeted therapy, antago-
nists of human CD28 are currently unavailable. So far, all antibodies
described as reacting against human CD28 are agonists. Indeed, CD28 is
a homodimeric receptor6whose degree of cross-linking is implicated in
signal transduction through its association with phosphatidylinositol
3-kinase (PI3-kinase) via the cytoplasmic domain.7However, monova-
lent fragments can inhibit CD28/B7 interactions without stimulating
CD28,7although they cannot be used therapeutically in vivo because of
their rapid elimination from the body. Extension of the half-life of
antibody fragments has been achieved by in vitro conjugation to one or
more molecules of polyethylene glycol8,9 or by molecular fusion with
serum albumin.10 Our group has recently investigated strategies aimed at
developing monovalent anti-CD28 reagents by cloning antigen-
combining portions and fusing them with monovalent carrier molecules.
In this paper we demonstrate that an scFv recombinant fragment from a
monoclonal antibody (mAb) directed against human CD28 (CD28.3
mAb11) fused with the monovalent and long-lived human serum protein
1-antitrypsin (HaaT), retains the avidity and the biologic properties of
the original monovalent Fab antibody fragment. In addition, this genetic
fusion extends the serum half-life of the recombinant protein further
than that of HaaT.
Materials and methods
Cells and antibodies
Mouse antihuman CD28 hybridoma CD28.1 (IgG1), CD28.2 (IgG1),
CD28.3 (IgG1), CD28.4 (IgM), CD28.5 (IgG1), and CD28.6 (IgG2a) and
mouse L fibroblasts expressing human CD80 (LB7 cells) were kindly
provided by D. Olive (INSERM U119).11,12 CD28.1, CD28.2, CD28.3, and
From the Institut de Transplantation et de Recherche en Transplantation, Institut
National de la Sante´ et de la Recherche Me´dicale (INSERM) U437, Centre
Hospitalier Universitaire (CHU) Hotel Dieu, Nantes, France; INSERM U463, Institut
de Biologie, Nantes, France; and Sangstat Europe, Lyon, France.
Submitted August 13, 2002; accepted March 9, 2003. Prepublished online as
Blood First Edition Paper, March 20, 2003; DOI 10.1182/blood-2002-08-2480.
Supported in part by the “Fondation Progreffe,” by the Post-Genome Program,
grant 109 from the French government (Ministe`re de l’Education Nationale de
la Recherche et de la Technologie [MENRT]), and by the association “Vaincre
La Mucoviscidose.”
Reprints: Bernard Vanhove, ITERT, INSERM U437, CHU Hotel Dieu, 30 Bld
Jean Monnet, 44093 Nantes, France; e-mail: bvanhove@nantes.inserm.fr.
The publication costs of this article were defrayed in part by page charge
payment. Therefore, and solely to indicate this fact, this article is hereby
marked ‘‘advertisement’’ in accordance with 18 U.S.C. section 1734.
© 2003 by The American Society of Hematology
564 BLOOD, 15 JULY 2003 VOLUME 102, NUMBER 2
For personal use only. by guest on June 12, 2013. bloodjournal.hematologylibrary.orgFrom
CD28.5 mAbs share a common target epitope, whereas CD28.4 and
CD28.6 mAbs bind to 2 other epitopes.11 Fab fragments were prepared
using the Immunopure IgG1 Fab preparation kit (Pierce, Rockford, IL).
In vivo infusion and blood sampling in macacus fascicularis
This part of the study was performed by Biomatech (Lyon, France) in
Cynomolgus maccaca fascicularis. Two infusions of 8 mg/kg antibodies
were performed on days 0 and 2.
Amplication and assembly of VHand VLby PCR
mRNA from CD28.3 hybridoma was prepared and retrotranscribed, and
cDNA for VHand VLwas amplied using primers for mouse V-gene
amplication of the mouse ScFv module of the recombinant phage antibody
system (Pharmacia Biotech, Guyancourt, France), according to the manufac-
turers instructions. The amplied V and H genes were cloned into a TA
vector (Invitrogen, Cergy Pontoise, France), followed by DNA sequencing.
The cloned VH gene was modied by polymerase chain reaction (PCR)
amplication by introducing a 5BamHI site (primer sc28.3-5: 5-
ATATGGATCCGTCAAGCTGCAGCAGTCAGG-3)anda3(SGGGG)3
synthetic linker primer (primer: 5-GACTGGGTCATCTGGATGTCCGA-
TCCGCCACCGCCAGAGCCACCTCCGCCTGAACCGCCTCCACC-
TGAGGAGACGGTGACCATGG-3). The VL gene was then assembled to
the modied VH gene by PCR using a 3primer (primer sc28.3-3:
5-ATATCTCGAGTTATTAGAATTCCCGTTTTATTTCCAGCTTGG-3)
introducing EcoRI and XhoI cloning sites and using the modied VH gene
as the 5primer. Assembled VH and VL genes were then reamplied using
a3primer introducing a TVAAPS peptidic linker originating from the
natural Fab hinge region of immunoglobulins and EcoRI and XhoI restric-
tion sites (primer sc28.3-TV5: 5-ATATCTCGAGTTATTAGAATTCA-
GATGGTGCAGCCACAGTCCGTTTTATTTCCAGCTT-3). The con-
struct was then sequenced to verify its identity with the original VH and VL
genes. The construct was further subcloned into the BamHI/XhoI sites of
the pSecTag2B eukaryotic expression plasmid (Invitrogen), 3to the Ig
leader and introduced into the EcoRI/EcoRV sites of the pIg6 prokaryotic
expression plasmid (a gift from Dr Jean-Charles Tellier, University of
Nantes, Nantes, France) 3to the OmpA leader.
Genetic fusion of scFv with HaaT
The cDNA for human 1-antitrypsin (corresponding to the GenBank
sequence no. X01683) was a gift from Dr D. Favre (INSERM ERM-0105,
Nantes, France). Amino acids 48 to 425 were amplied by PCR with
primers that add anking EcoRI restriction sites (5primer: 5-
ATATGAATTCAACAAGATCACCCCCAAC-3;3primer: 5-ATAT-
GAATTCTTTTTGGGTGGGATTCAC-3). The modied gene was in-
serted into the EcoRI site of the pSecTag2B-scFv28.3, 3and into the
EcoRVsite of the pIg6-scFv28.3 in frame with the scFv cDNA.
Expression in Escherichia coli
JM83 cells were transformed with the pIg6-scFv28.3-HaaT plasmid.
Transfectants were allowed to grow in Luria-Bertani (LB) medium
containing 100 g/mL ampicillin until an OD600 of 0.5 was reached. Then,
1 mM isopropyl thiogalacto-pyranoside (IPTG) was added to the cultures
and the cells were incubated at 25°C for 3 hours. The cells were then
centrifuged and the periplasmic extracts were collected after hypotonic
lysis and ultracentrifugation at 10 000g.
Transient transfection of Cos cells and purication
of scFv28.3-HaaT
Cos cells were transfected using the Fugene lipofection kit (Roche
Diagnostics, Basel, Switzerland) according to the manufacturers instruc-
tions. Cultures were maintained for 3 days at 37°C, divided one third, and
put back into culture for an additional 3 days, after which time the medium
was collected. Supernatants were passed through NiNTA Sepharose col-
umns (Amersham Pharmacia Biotech, Saclay, France) at a rate of
0.5 mL/min. The columns were rinsed with 0.02 M imidazole in phosphate-
buffered saline (PBS) and proteins were eluted with 0.2 M imidazole in
PBS and then dialyzed extensively against PBS at 4°C.
Enzyme-linked immunosorbent assay
Recombinant human CD28-Fc (R&D Systems, Abingdon, United King-
dom) was used at 1 g/mL in borate buffer (pH 9.0) to coat 96-well
microtiter plates (Immulon, Chantilly, VA) overnight at 4°C. Reactive sites
were blocked with 5% skimmed milk in PBS for 2 hours at 37°C and
supernatants or puried material were reacted for 2 hours at 37°C. Bound
scFV28.3-HaaT was revealed with successive incubations (1 hour, 37°C)
with rabbit antihuman 1-antitrypsin antibodies (1:500 dilution; Dako,
Trappes, France) and horseradish peroxidase (HRP)conjugated donkey
antirabbit Ig antibodies (1:500 dilution; Jackson ImmunoResearch Labora-
tories, Bar Harbor, ME). Control scFv-M13 was revealed with an HRP-
conjugated anti-M13 antibody (Amersham Pharmacia Biotech), as de-
scribed.13 Bound antibody was revealed by colorimetry using the ABTS
substrate (Roche, Mannheim, Germany) read at 405 nm.
EliSpot assay
CD4T cells (105) were mixed with irradiated allogeneic peripheral blood
mononuclear cells (PBMCs; 105) in RPMI 1640 medium (Sigma, St
Quentin Fallavier, France) supplemented with glutamine, nonessential
amino acids, sodium pyruvate, antibiotics, and 10% heat-inactivated fetal
calf serum (FCS) and plated in quadruplicate into microtiter plates coated
with antiinterferon-(antiIFN-) mAb (AID, Strassberg, Germany).
After a 24-hour incubation at 37°C, wells were washed, incubated with a
biotin-labeled secondary antiIFN-mAb, and revealed according to the
manufacturers instructions. Spots were counted with an EliSpot Reader
System (AID).
Detection of scFv28.3-HaaT by Western blotting
scFv28.3-HaaT fusion proteins were electrophoresed in 10% polyacryl-
amide gels and blotted onto nitrocellulose membranes. Blots were revealed
with an antic-myc mAb (prepared in our laboratory from 9E10 hybridoma)
and an HRP-conjugated donkey antimouse Ig antibody (Jackson Immuno-
Research Laboratories) and developed by chemiluminescence (Amersham
Pharmacia Biotech).
Adhesion assay
Adhesion assays were performed as previously described12 except that
Jurkat T cells were loaded with calcein AM (Molecular Probes, Eugene, OR)
instead of being radiolabeled. Jurkat T cells (6 104) were added to a
monolayer of 104LB7 cells (expressing human CD80) and incubated at
37°C for 1 hour.Adherent cells were analyzed by uorometry (excitation at
485 nm and emission at 430 nm).
Cytouorometry
Jurkat T cells or U937 cells were incubated for 1 hour at 4°C with
supernatants from control or transfected Cos cells, or with puried proteins.
Bound scFv28.3-HaaT fusion proteins were detected with a rabbit antihu-
man 1-antitrypsin antibody (1:200 dilution; Dako) and a uorescein
isothiocyanate (FITC)conjugated donkey antirabbit Ig antibody (dilution
1:200; Jackson ImmunoResearch Laboratories) for 30 minutes at 4°C. Cells
were then analyzed by uorescence-activated cell sorting (FACS). Monkey
PBMCs were prepared by centrifugation of blood on Ficoll (Pharmacia
Biotech) and incubated on ice in PBS/0.1% NaN3with a mixture of
antiCD2-phycoerythrin (PE) mAb (Immunotech, Marseille, France) and
FITC-labeled anti-CD28.6 mAb (prepared in our laboratory) for revelation
of CD28 on T cells from monkeys treated with CD28.1, CD28.2, CD28.3,
and with a mixture of antiCD2-PE plus FITC-labeled anti-CD28.2 mAb
(prepared in our laboratory), for revelation of CD28 on T cells from
monkeys treated with CD28.6.
Mixed lymphocyte reactions
Human PBMCs were seeded in triplicate at a nal concentration of
105cells/well in RPMI 1640 medium (Sigma) supplemented with glu-
tamine, nonessential amino acids, sodium pyruvate, antibiotics, and
ANTI-CD28 scFv1-ANTITRYPSIN FUSION ANTIBODY 565
BLOOD, 15 JULY 2003 VOLUME 102, NUMBER 2
For personal use only. by guest on June 12, 2013. bloodjournal.hematologylibrary.orgFrom
10% heat-inactivated FCS and cultured for 5 days with 105allogeneic
PBMCs irradiated at 30 Gy. Proliferation was measured by 3H-thymidine
incorporation after 16 hours of incubation with 106Ci (37 KBq).
Alternatively, CD4T cells prepared from PBMCs using the Lymph-Kwik
Th system (One Lambda, Canoga Park, CA) were used as responding cells.
The results were expressed as specic counts per minute per triplicate: (cpm
assay cpm stimulating cells alone cpm responding cells alone).
Capping and immunouorescence microscopy
Capping experiments were performed using a modied procedure of
Gassmann et al.14 Jurkat T cells were incubated either at 37°C for 1 hour
with mouse mAb CD28.3 (10 g/mL), CD28.3 Fab fragments, scFv28.3-
HaaT, or scFv28.3 recombinant proteins in RPMI 1640 medium (Sigma)
containing 10% FCS, or at 0°C for 1 hour in the same medium containing
0.1% NaN3. After washing, cells were immediately xed in 0.5%
paraformaldeyde (wt/vol) for 30 minutes at room temperature, centrifuged
onto glass slides, and allowed to dry overnight. Slides were immunolabeled
with either FITC-labeled goat antimouse IgG antibodies (Jackson Immu-
noResearch Laboratories), rabbit antihuman 1-antitrypsin followed by
FITC-labeled donkey antirabbit IgG antibodies, or mouse antic-myc 9E10
mAb followed by goat antimouse IgG antibodies. After washing, the slides
were mounted in Moviol and examined using the 63 immersion lens of a
Zeiss microscope equipped for epiuorescence.
Biosensor afnity measurement
CD28-Fc recombinant protein (R&D Systems) was immobilized onto a
CM5 sensor chip. Analysis was performed with a BIAcore 2000 (Biacore,
Paris, France). Antibodies or recombinant proteins were applied at concen-
trations ranging from 0.67 to 12.5 nM with a ow rate of 30 L/min. An
association period of 300 seconds was followed by a dissociation period of
600 seconds. Analyses and binding constants were deduced using the
BIAevaluation software (Biacore).
Pharmacokinetic analysis of the conjugate
Proteins (30-60 g) were labeled with 100 g iodogen and 3.106Ci (110
KBq)/mg 125I as described.15 Free iodine was eliminated by chromatogra-
phy on Sephadex G-25M using a PD10 column (Amersham Pharmacia
Biotech). Via thin layer chromatography with 10% trichloroacetic acid it
was determined that 91% to 96% of the radioactivity was associated with
the proteins. The specic activity of each preparation was calculated from
estimates of protein concentration (measured by absorption at 280 nm) and
radioactivity and was typically in the range of 0.1 to 0.3 106Ci (4 to 10
KBq)/g. The radiolabeled samples were used directly after labeling.
Thirty or 60 g125I-labeled protein in PBS was injected into the tail vein of
10-week-old male Balb/c mice. Blood samples were collected periodically
from the orbital sinus. Blood samples were weighed and radioactivity
detected using a gamma counter (Canberra Packard Topcount, Mississauga,
ON, Canada). Percent injected dose (%ID) was calculated for each
individual mouse and expressed as %ID/mL total blood volume. The data
were analyzed by Siphar software (Simed, Utrecht, The Netherlands) with
the use of a 2-compartment model.
Results
The antihuman CD28 mAbs tested do not down-regulate
their target in vivo
In the rat species, one mAb is known to induce CD28 capping and
complete down-regulation in vivo.16 Thus, we rst screened a
series of antihuman CD28 mAbs for cross-reaction with macaques
to investigate a possible down-regulating effect in vivo. Cross-
reacting mAbs (CD28.1, CD28.2, CD28.3, and CD28.6 mAb12)
were then injected intravenously into macacus fascicularis and
CD28CD2cell counts and CD28 expression intensity were
monitored in the blood (Figure 1). No reduction in CD28 expres-
sion intensity was noted. Conversely, it was increased, together
with an increase in the mean forward scatter (FSC) signal in ow
cytometry (not shown). CD28cell counts were unchanged
(CD28.3) or reduced by 30% for CD28.2, 50% for CD28.1, and
60% for CD28.6.
Selection of an anti-CD28 Fab fragment inhibiting
CD28/B7 interaction
In the absence of modulating activity of the anti-CD28 antibodies
tested, we screened the CD28 blocking ability of their monovalent
fragments. Fab fragments were prepared and tested by ow
cytometry, inhibition of adhesion of Jurkat T cells to CD80-
transfected broblasts, and inhibition of mixed lymphocyte reac-
tions (MLRs; Figure 2). Reactivity was compared with initial
divalent IgG molecules. As determined by FACS, the CD28.1 and
CD28.2 antibodies reduced their binding activity at least 100-fold
after digestion into monovalent Fab fragments. In contrast, the
reduction in reactivity of Fab fragments from CD28.3 was below a
factor of 10 (Figure 2A). An evaluation of binding parameters
performed by Biacore conrmed that Fab monovalent fragments
from the CD28.3 mAb had a higher association rate (Ka, 9650/
mole-second) than Fab fragments from the CD28.2 and CD28.1
mAb (Ka, 920/mole-second and below the detection limit, respec-
tively). Fab fragments from the CD28.3 antibody fully inhibited the
adhesion of CD28T cells to CD80-expressing cells (Figure 2B),
whereas fragments from the other antibodies only partially reduced
or had no effect on the adhesion of T cells. In MLRs, anti-CD28
antibodies are expected to costimulate (cross-linking of CD28) or,
alternatively, to reduce T-cell proliferation (CD28/B7 interaction
inhibition). In contrast, monovalent fragments are expected to
inhibit T-cell proliferation. The anti-CD28 antibodies in their
dimeric conguration showed no inhibition of proliferation in
MLRs and some of them promoted T-cell proliferation at high
doses (CD28.1, Figure 2C). Monovalent Fab fragments from the
CD28.3 antibody showed a dose-response inhibition of prolifera-
tion, whereas Fab fragments from the other antibodies did not,
probably as a result of their lower binding avidity. Finally,
observations by immunouorescence revealed that the divalent
anti-CD28 mAb CD28.3 induced a capping of CD28 molecules
on T-cell surfaces at 37°C, whereas monovalent Fab fragments
did not.
Construction and expression of sc28.3HaaT from
the CD28.3 hybridoma
VHand VLchain fragments were amplied from CD28.3 hybrid-
oma cDNA and assembled using a (GGGGS)4linker. The nucleo-
tide sequence of this scFv was registered in GenBank under the
Figure 1. Assessment of a possible in vivo modulating activity of anti-CD28
antibodies. Anti-CD28 mAb (8 mg/kg) was infused intravenously into macacus fascicu-
laris on days 0 and 2. Blood samples were collected before injection and every 2 days for
10 days. PBMCs were then analyzed by ow cytometry after gating of CD2cells. Mean
uorescence intensity (MFI) of CD28 expression (A) as well as percent of CD2CD28
double-positive cells within mononuclear cells (B) are represented; indicates CD28.1; Œ,
CD28.2; f, CD28.3; F, CD28.6; and E, isotype control. Similar data were found in 2
monkeys per antibody, one of them being shown here.
566 VANHOVE et al BLOOD, 15 JULY 2003 VOLUME 102, NUMBER 2
For personal use only. by guest on June 12, 2013. bloodjournal.hematologylibrary.orgFrom
accession no. AF451974. To load the scFv with a carrier molecule
that would increase the molecular weight of the complex and give
an extended bioavailability in vivo, we selected a series of
monovalent macromolecules that are abundant in plasma. We rst
fused the coding sequence of serum albumin to the C-terminus of
the scFv because this had already been used successfully in a rabbit
system.17,18 In the meantime, feasibility was reported with human
albumin.10 In our hands, the genetic fusion of scFv 28.3 with serum
albumin resulted in the production of a misfolded, insoluble
complex without antibody activity in prokaryotic and eukaryotic
expression systems (not shown). In contrast, the genetic fusion of
scFv28.3 with a TVAAPS Fab hinge sequence and with amino
acids 53 to 425 (referring to GenBank accession no. K01396) of the
human 1-antitrypsin resulted in the production of a soluble and
active molecule, as illustrated in Figure 3. The scFv28.3-HaaT
cDNA was rst subcloned into the EcoRV/XhoI sites of the pIg6
prokaryotic expression plasmid (a generous gift from Dr Jean-
Charles Tellier, University of Nantes, Nantes, France) 3to an
ompA signal sequence, enabling accumulation of recombinant
proteins in the periplasm of JM83 E coli cells, under the control of
the lac promoter (Figure 4A). However, periplasmic extracts
contained only low levels of soluble protein. The scFv28.3-HaaT
cDNA was subsequently subcloned into the BamHI/EcoRI sites of
the pSecTag2B pCMV-based eukaryotic expression plasmid, en-
abling a fusion at the C-terminus with the Igleader sequence and
at the N-terminus with a 6-HIS tag. After transfection in Cos cells,
anti-CD28 activity was found in the supernatant and the recombi-
nant protein was puried on NiNTAafnity columns with a yield of
1.5 mg/L supernatant (Figure 4B-D).
Detection of sc28.3HaaT binding activity by ow cytometry,
ELISA, and biosensor
The binding of the scFv28.3-HaaT construct was then analyzed by
ow cytometry using CD28Jurkat T cells and control U937 cells.
The binding activity was tested by adding the scFv28.3-HaaT
construct to Jurkat cells followed by staining with a rabbit
anti1-antitrypsin antibody and an FITC-conjugated donkey anti-
rabbit Ig antibody as a third-step reagent and analysis by ow
cytometry. The results indicated that scFv28.3-HaaTbound speci-
cally to CD28 expressed on Jurkat cells with no binding to U937
cells, which do not express CD28 (Figure 4). Binding was also
conrmed in an enzyme-linked immunosorbent assay (ELISA),
performed by adding various concentrations (25-400 ng/mL) of
scFv28.3-HaaT to plates coated with recombinant CD28-Fc mol-
ecules (Figure 4C). Negative controls where plates were coated
with irrelevant molecules (gelatin, bovine serum albumin, ovalbu-
min) gave no signal (not shown). In addition, an unrelated scFv
(fused with the G3p protein from M13 bacteriophage) and 1-
antitrypsin alone did not bind to recombinant CD28-Fc. From this
analysis, the inhibitory concentration of 50% (IC50) was estimated
as being 0.4 nM. The avidity of the scFv28.3-HaaT was evaluated
via a biosensor analysis. Adetector chip was coated with CD28-Fc
Figure 2. Characterization of the Fab fragments of anti-CD28 antibodies.
(A) CD28Jurkat T cells were incubated with the indicated amounts of anti-CD28
mAb or their Fab fragments and stained with antimouse-FITC antibody. MFI was
determined by ow cytometry. (B) Inhibition of CD28/CD80 interactions was as-
sessed by incubating calcein-labeled CD28Jurkat T cells on monolayers of mouse
broblasts expressing human CD80 in the presence of saturating doses of anti-CD28
Fab fragments or control Fab fragments. After washing, adherent cells were collected
and associated uorescence was measured by uorometry. (C) PBMCs (105) were
mixed with 105allogeneic irradiated PBMCs in the presence of the indicated amount
of anti-CD28 mAb and Fab fragments or with 10 g/mL control antibody. Proliferation
on day 5 was measured by addition of 106Ci (37 KBq) 3H-thymidine and
measurement of incorporation after 16 hours; indicates IgG CD28.1; Œ, IgG
CD28.2; f, IgG CD28.3; , Fab CD28.1; , Fab CD28.2; , Fab CD28.3; and ,
isotype control. The results shown are representative of more than 3 independent
experiments.
Figure 3. Construction for expression of scFv28.3-HaaT in eukaryotic cells.
cDNA fragments coding for variable regions of VHand VLchains of the CD28.3
antibody were assembled with a (GGGGS)4linker. AcDNA coding for the TVAAPS hinge
peptide from the IgG heavy chain was added 3and fused in frame with a cDNA coding for
amino acids 53 to 425 of HaaT cDNA. The complex was then introduced into the
pSecTag2B expression plasmid by adding an Igleader to the N-terminus and a stretch
sequence containing c-myc and 6HIS ag plus a stop codon to the C-terminus.
Figure 4. Expression and activity of scFv28.3-HaaT. (A) Prokaryotic expression:
10 g protein extract from the periplasmic, soluble (S), and insoluble (I) fractions of
E coli JM83 expressing scFv28.3-HaaT were resolved under reducing conditions on 10%
polyacrylamide electrophoresis gels and blotted onto nylon membranes. Recombinant
proteins were revealed by incubation of the membranes with HRP-conjugated antic-myc
antibody and by enhanced chemiluminescence (ECL). (B) Fifty nanograms scFv28.3-
HaaT produced in Cos cells, under native (N) or reducing (R) conditions, was analyzed by
Western blotting as described in panel A. Molecular size markers (kDa) are shown on the
left. (C) CD28Jurkat T cells were incubated with supernatant from transfected Cos cells
containing 2 g/mL scFv28.3-HaaT (black histogram, rst row) or with supernatant from
control Cos cells (white histogram, second row), revealed with anti-HaaT antibody plus
FITC-conjugated donkey antirabbit Ig antibody (DAR) and analyzed by cytouorometry.
CD28U937 cells were analyzed in parallel with supernatant containing scFv28.3-HaaT
(dark gray histogram, third row) or with control supernatant (light gray histogram, last row).
(D) The reactivity of Cos-produced scFv28.3-HaaT was examined by ELISA. CD28-Fc
molecules were coated onto microtiter plates. Puried scFv28.3-HaaT at different dilutions
in PBS-Tween was added and incubated for 1 hour at 37°C. After washing, bound
scFv28.3-HaaT molecules were revealed with a rabbit anti-HaaT antibody followed by a
peroxidase-conjugated DAR as a third-step reagent. Colorimetric analysis was performed
after reaction of ABTS with peroxidase. X indicates signal without sc28.3-HaaT; and ,
signals obtained with incubations of HaaT alone and with an irrelevant scFv-M13 fusion
antibody, revealed with a peroxidase-labeled anti-M13 antibody. Data are presented as
means of triplicates.
ANTI-CD28 scFv1-ANTITRYPSIN FUSION ANTIBODY 567
BLOOD, 15 JULY 2003 VOLUME 102, NUMBER 2
For personal use only. by guest on June 12, 2013. bloodjournal.hematologylibrary.orgFrom
at high and low densities. Recombinant proteins were applied at
concentrations ranging from 0.67 to 12.5 nM with a ow rate of 30
L/min, an association period of 300 seconds followed by a
dissociation period of 600 seconds. The analysis of the avidity,
deduced from the BIA evaluation software from 6 determinations
at different doses, revealed a Kd(M) of 7.1 1.8 109. Similar
values were obtained in 3 independent analyses using either Cos
cell supernatants or puried material. A similar analysis of the
initial CD28.3 Fab fragments revealed avidity in the same order of
magnitude (Kdof 8.5 109).
Detection of sc28.3HaaT aggregation by Western blotting
and absence of capping induction
Recombinant scFv antibodies often tend to aggregate or form
dimers in solution19; thus, even after the fusion with a monovalent
carrier such as HaaT, our scFv28.3 may dimerize and induce
clustering of CD28 molecules on T-cell membranes. To character-
ize the nature of the recombinant protein produced in Cos cells,
supernatants were analyzed by electrophoresis under native and
denaturant conditions. After reduction and denaturation in sodium
dodecyl sulfate (SDS), the protein had an apparent molecular mass
of 90 kDa (Figure 4B). The fact that the same protein produced in
E coli had an apparent molecular mass of 78 kDa (Figure 4A)
indicated that approximately 12 kDa could be attributed to
glycosylation after production in eukaryotic cells. Under native
conditions, without SDS, the protein was mostly monomeric with
an apparent molecular weight similar to that of the denatured
protein. Material with a molecular weight of approximately 180
kDa was also detected, indicating the presence in the solution of
dimeric proteins (Figure 4). The capping assay, however, revealed
that the scFv28.3-HaaT produced in Cos cells, unlike divalent IgG
molecules, did not induce formation of CD28 clusters on T-cell
membranes, and neither did an scFv28.3 nonfused to the HaaT
(Figure 5). This suggests that the dimeric proteins contained in the
preparation could not stimulate CD28 either due to their associa-
tion being too weak or due to competition for binding with
quantitatively dominant monovalent fragments.
scFv28.3-HaaT modulates T-cell allogeneic responses
Our group20 has previously demonstrated that monovalent Fab
fragments from CD28 antibodies reduce proliferation of alloreac-
tive T cells but are ineffective in modulating proliferation of Tcells
stimulated by heterologous antigens presented in the context of self
major histocompatibility complex (MHC) molecules. To investi-
gate whether scFv28.3-HaaT could similarly reduce T-cell allore-
sponses, we rst checked whether this construct inhibited CD28/B7
interactions and inhibited an MLR. As shown in Figure 6,
scFv28.3-HaaT dose dependently inhibited adhesion of Jurkat T
cells to CD80-expressing broblasts (Figure 6A) and reduced
proliferation of allogeneic T cells in vitro (Figure 6B). CD28
blockade also reduced the frequency of IFN-producing cells in
MLRs 24 hours after stimulation (Figure 6E).
To determine whether the reduced alloreactivity in MLRs with
CD28 blockade was due to immune ignorance, anergy, or deletion
of alloreactive T cells, we measured secondary responses of cells
initially primed in the presence of sc28.3-HaaT. No modications
in the frequency of IFN-secreting cells (Figure 6E) or in
proliferation (Figure 6C) were noted after secondary stimulation,
indicating that alloreactive cells had not been deleted in the primary
stimulation. Inhibiting CD28 in primary MLRs with Fab fragments
from the CD28.3 antibody instead of sc28.3HaaT led to similar
conclusions (data not shown). However, proliferation in recall
stimulation was maximal on day 2, whereas it peaked on day 3
when third-party stimulating PBMCs were used (Figure 6D),
indicating that donor-specic proliferating cells had been primed in
the primary reaction despite CD28 blockade.
Pharmacokinetics in murine plasma
To address the question of whether the genetic fusion of the scFv
fragment with HaaT actually resulted in an extended half-life in
vivo, we followed the distribution in mice of iodinated scFv28.3-
HaaT, Fab fragments from the CD28.3 antibody, and HaaT. Figure 7
shows that the distribution and elimination half-lives of the Fab
fragments were rapid, as expected for a 50-kDa protein (14.5 3.3
minutes and 7.4 1.7 hours, respectively). HaaT distribution was
similar (17.4 10.8 minutes) but it was eliminated much more
slowly (10.8 0.6 hours) despite having a slightly lower molecu-
lar weight (46.6 kDa). The distribution and the elimination
half-lives were signicantly extended for the scFv28.3-HaaT
recombinant molecule (26.6 2.3 minutes and 11.8 0.5 hours,
respectively). Finally, an analysis of the biodistribution of the
labeled molecules 48 hours after injection conrmed the accumula-
tion of scFv28.3-HaaT in the reticuloendothelial compartment
(liver, kidneys, intestine, spleen; not shown), whereas Fab frag-
ments were lost, probably through renal elimination. Furthermore,
the distribution volume increased from 3 mL for Fab and HaaT to 8
mL for scFv28.3-HaaT, indicating that the conjugate left the blood
to penetrate organs and tissues more rapidly.
Discussion
Antibody-induced modulation of CD28 prolongs allograft survival
in rats16 and administration of anti-CD28 Fab fragments reverses
the induction of experimental autoimmune encephalomyelitis21 and
uveoretinitis22 in mice. In a murine model of graft-versus-host
Figure 5. Immunouorescent analysis of antibody-induced capping of CD28.
Jurkat T cells expressing CD28 were incubated in culture medium with 2 g/mL
divalent CD28.3 IgG antibody (A-B) or 2 g/mL CD28.3 Fab fragments, with 2 g/mL
scFv28.3-HaaT (E) or with 50% Cos cell supernatant containing approximately
4g/mL scFv28.3 (D). Cells were incubated for 1 hour at 0°C (A) or at 37°C (B-E),
washed in ice-cooled PBS containing NaN3, and centrifuged onto microscope slides.
After xation with 0.5% paraformaldehyde (PFA), CD28 molecules were revealed
using FITC-labeled CD28.6, a mAb reacting against an epitope on CD28 other than
CD28.3.11 Slides were observed by uorescence microscopy, and magnication is
63 for all panels.
568 VANHOVE et al BLOOD, 15 JULY 2003 VOLUME 102, NUMBER 2
For personal use only. by guest on June 12, 2013. bloodjournal.hematologylibrary.orgFrom
disease (GVHD), the selective blockade of CD28 was reported as
being more immunosuppressive than blocking B7 with CTLA-4/Ig.23
CD28 blockade also synergizes with cyclosporine in the rat
resulting in immune tolerance in transplantation.24 In CD28/
mice, immune responses to viral antigens or autoantigens are
impaired,25,26 whereas responses to exogenous antigens remain
normal.27 In rats treated with the modulating anti-CD28 antibody
JJ319, allogeneic T-cell responses are blunted but responses to
exogenous antigens are unaffected.20 Collectively, these data
suggest that T-cell responses to autoantigens or alloantigens are
more dependent on costimulation through CD28 than T-cell
responses to exogenous antigens. Thus, the blockade of CD28 may
represent an immunosuppression for the selective inhibition of
pathologic T cells in autoimmunity and in transplantation without
inhibition of other, protective, T-cell responses.
Costimulation through CD28 in conjunction with a triggering of
the T-cell receptor (TCR) leads to high-level production of
interleukin 2 (IL-2) and provides an essential survival signal for T
cells. Signals through CD28 also regulate T-cell cycle entry and
progression through the G1phase in an IL-2independent man-
ner,28 resulting in the activation of cyclines.29 At the molecular
level, a selective blockade of CD28 with an unmodied CTLA-
4/B7 interaction would theoretically allow signals through the TCR
and CTLA-4 to be transmitted to antigen-challenged T cells.
Resting T cells express a relatively low level of CTLA-4; however,
when engaged on resting T cells, CTLA-4 transmits a signal that
blocks transition from G0to G1.30 Once activated, T cells further
increase their membrane expression of CTLA-4, which transmits a
signal leading to a Fas-independent cell death,31 contributing to the
down-regulation of the immune response. Therefore, allowing
CTLA-4/B7 interactions to proceed in the absence of CD28
costimulation will combine the effect of the lack of costimulation
with a cell growth arrest, or a clonal deletion, depending on
whether blockade is initiated before or after the initiation of T-cell
activation. In addition, as agonistic anti-CD28 antibodies promote
Th1 differentiation in vitro,32 inhibition of CD28 is likely to modify
the Th1/Th2 balance.
This study was undertaken to obtain an anti-CD28 antagonist
that would allow an in vivo investigation into whether recognition
of alloantigens or autoantigens by CD4T cells in the absence of
CD28 costimulation, but with an unmodied CTLA-4/B7 interac-
tion, induces an immune regulation. The available anti-CD28
antibodies appear not to be antagonistic in vivo in monkeys, as
shown by the absence of CD28 down-regulation. On the contrary, 3
of the 4 antibodies we tested induced a strong up-regulation of
CD28 expression on CD2cells (Figure 1), a phenomenon that has
previously been associated with T-cell activation.33
In the absence of identied modulating anti-CD28 antibodies,
we constructed a recombinant monovalent antibody from the
hybridoma of CD28.3. This clone was chosen because (1) it retains
its high afnity even after digestion into monovalent Fab frag-
ments, (2) it prevents CD28 from binding CD80, (3) its Fab
fragments dose dependently reduce T-cell proliferation in MLRs,
and (4) it retains its binding properties after recombinant expres-
sion in scFv format. The extension of its molecular weight through
a genetic fusion with 1-antitrypsin resulted in a soluble macromol-
ecule with an extended bioavailability and a preserved activity.
1-Antitrypsin is an abundant (3 g/L) monovalent plasma protein
that has already been used for genetic fusion.34 In humans it has a
half-life of about 8 days,35 although its half-life is much shorter in
rodents.36 To demonstrate the immunosuppressive potential of
scFv28.3-HaaT, we reproduced the biologic activities of the
CD28.3 antibody Fab fragments in vitro. This included a preserved
avidity for CD28, the absence of induction of CD28 capping on
target cells at 37°C, a competition with CD80 for binding to CD28,
and a dose-dependent inhibition in MLRs. In addition, sc28.3HaaT
reduced the frequency of allogeneic cells producing IFN-24 hours
after the initiation of an MLR.
Figure 6. Biologic activity of scFv28.3-HaaT in a binding assay and MLRs.
(A) CD28Jurkat T cells stained with calcein AM dye were incubated on a monolayer
of CD80-expressing broblasts, as in Figure 2B, in the presence of dilutions of
supernatant from Cos cells transfected with pSecTag2B-scFv28.3-HaaT (f)or
pSecTag2B-scFv28.3 (), in which recombinant protein was quantied by ELISA.
Control-transfected supernatant (E) was used at similar dilutions. The percentage of
inhibition of adhesion after washing was evaluated by luminescence. (B) CD4T cells
from healthy volunteers were incubated in microtiter plates with irradiated allogeneic
PBMCs for 5 days in the presence of dilutions of puried sc28.3-HaaT. Proliferation was
measured by incorporation of 3H-thymidine for the last 16 hours of culture; m indicates
proliferation obtained after addition of a control mAb (mouse IgG1) to the cultures. Data
shown here are means SDs of triplicates and are representative of 5 experiments.
(C-E) MLRs similar to panel B were performed with (lled bars and symbols) or without
(empty bars and symbols) saturating amounts of sc28.3-HaaT.After 5 days an aliquot of
cells was assessed for proliferation (primaryin panel C) and for frequency of IFN-
secretion (primaryin panel E). The remaining cells were washed and cultured for a resting
period of 7 days, after which time cells were collected and restimulated (secondary) with
the same (C, circles in panels D and E) or third-party (triangles in panel D) stimulatory cells.
Proliferation was measured 2 (C-D), 3, and 4 (D) days later. The frequency of IFN-
secreting cells during secondary stimulation was measured 24 hours after restimulation
(secondaryin panel E).
Figure 7. Pharmacokinetics of 125I-labeled HaaT, CD28.3Ab Fab fragments, and
scFv28.3-HaaT in mice. Puried sc28.3-HaaT, HaaT, and CD28.3 Fab fragments
were labeled with 125I and injected intravenously into mice. Blood samples were
collected periodically and radioactivity was measured with a gamma counter. %ID
indicates percent injected dose; f, scFv28.3-HaaT; E, HaaT; and , Fab. For each,
n3. Data are presented as means SDs.
ANTI-CD28 scFv1-ANTITRYPSIN FUSION ANTIBODY 569
BLOOD, 15 JULY 2003 VOLUME 102, NUMBER 2
For personal use only. by guest on June 12, 2013. bloodjournal.hematologylibrary.orgFrom
Restimulation of T cells primed in the presence of the CD28
inhibitor resulted in intact secondary responses, suggesting that one
mechanism of action of sc28.3-HaaT in MLRs that results in an
inhibition of T-cell proliferation and activation is related to immune
ignorance rather than anergy or alloreactive cell deletion. However,
the earlier kinetics of T-cell proliferation following stimulation by
isogeneic antigen-presenting cells (APCs) as compared with third-
party APCs in secondary responses (proliferation peak on day 2
instead of day 3) suggests that T cells were primed in the primary
reaction whether or not CD28 had been blocked. Additional
mechanisms to ignorance might, however, play a role in vivo
because a short-course administration of Fab fragments or of a
modulating anti-CD28 mAb in rodents increased apoptosis of
alloreactive cells20 and, in the long term, improved disorders
related to alloreactivity23 and autoimmunity.21,22 Thus, deletion of
alloreactive cells or the development of regulatory mechanisms
might also result from selective CD28 blockade. The possibility
that a selective blockade of CD28 in humans could improve
disorders such as GVHD or allograft survival, however, remains to
be demonstrated.
In summary, we have produced a reagent that, for the rst time,
can specically inhibit CD28/B7 interactions without interfering
with those of CTLA-4/B7. This agent is efcient at inhibiting
several CD28-mediated processes in vitro. We believe that this new
strategy may have clinical applications in humans.
Acknowledgments
The authors thank J. Naulet and J. Lafond for assistance in protein
production and purication, S. Iyer for the preparation of Fab fragments,
and Alain Faivre-Chauvet for assistance in protein iodination.
References
1. Schwartz RS. The new immunologythe end of
immunosuppressive drug therapy? N Engl J Med.
1999;340:1754-1756.
2. Alegre ML, Shiels H, Thompson CB, Gajewski TF.
Expression and function of CTLA-4 in Th1 and
Th2 cells. J Immunol. 1998;161:3347-3356.
3. Fallarino F, Fields PE, Gajewski TF. B71 en-
gagement of cytotoxic T lymphocyte antigen 4
inhibits T cell activation in the absence of CD28.
J Exp Med. 1998;188:205-210.
4. Kingsley CI, Karim M, Bushell AR, Wood KJ.
CD25CD4regulatory T cells prevent graft re-
jection: CTLA-4- and IL-10 dependent immuno-
regulation of alloresponses. J Immunol. 2002;
168:1081-1086.
5. Greenwald RJ, Boussiotis VA, Lorsbach RB, Ab-
bas AK, Sharpe AH. CTLA-4 regulates induction
of anergy in vivo. Immunity. 2001;14:145-155.
6. Williams AF, Barclay AN. The immunoglobulin
superfamilydomains for cell surface recogni-
tion. Annu Rev Immunol. 1988;6:381-405.
7. Damle NK, Doyle LV, Grosmaire LS, Ledbetter
JA. Differential regulatory signals delivered by
antibody binding to the CD28 (Tp44) molecule
during the activation of human T lymphocytes.
J Immunol. 1988;140:1753-1761.
8. Kitamura K, Takahashi T, Yamaguchi T, et al.
Chemical engineering of the monoclonal antibody
A7 by polyethylene glycol for targeting cancer
chemotherapy. Cancer Res. 1991;51:4310-4315.
9. Chapman AP, Antoniw P, Spitali M, West S, Ste-
phens S, King DJ. Therapeutic antibody frag-
ments with prolonged in vivo half-lives. Nat Bio-
technol. 1999;17:780-783.
10. Smith BJ, Popplewell A, Athwal D, et al. Pro-
longed in vivo residence times of antibody frag-
ments associated with albumin. Bioconjug Chem.
2001;12:750-756.
11. Nune` s J, Klasen S, Ragueneau M, et al. CD28
mAbs with distinct binding properties differ in their
ability to induce T cell activation: analysis of early
and late activation events. Int Immunol. 1993;5:
311-315.
12. Pages FRM, Klasen S, Battifora M, et al. Two dis-
tinct intracytoplasmic regions of the T-cell adhe-
sion molecule CD28 participate in phosphatidyl-
inositol 3-kinase association. J Biol Chem. 1996;
271:9403-9409.
13. Vanhove B, Charreau B, Cassard A, Pourcel C,
Soulillou JP. Intracellular expression in pig cells of
anti-alpha1,3galactosyltransferase single-chain
FV antibodies reduces Galalpha1,3Gal expres-
sion and inhibits cytotoxicity mediated by anti-Gal
xenoantibodies. Transplantation. 1998;66:1477-
1485.
14. Gassmann M, Guttinger M, Amrein KE, Burn P.
Protein tyrosine kinase p59fyn is associated with
the T cell receptor-CD3 complex in functional hu-
man lymphocytes. Eur J Immunol. 1992;22:283-
286.
15. Fraker PJ, Speck JC Jr. Protein and cell mem-
brane iodinations with a sparingly soluble chloro-
amide, 1,3,4,6-tetrachloro-3a,6a-diphrenylgly-
coluril. Biochem Biophys Res Commun. 1978;80:
849-857.
16. Dengler TJ, Szabo G, Sido B, et al. Prolonged
allograft survival but no tolerance induction by
modulating CD28 antibody JJ319 after high-
responder rat heart transplantation. Transplanta-
tion. 1999;67:392-398.
17. Makrides SC, Nygren PA, Andrews B, et al. Ex-
tended half-life of human soluble complement
receptor type 1 fused to a serum albumin-binding
receptor. J Pharmacol Exp Ther. 1996;277:534-
542.
18. Syed S, Schuyler PD, Kulczycky M, Shefeld WP.
Potent anti-thrombin activity and delayed clear-
ance from the circulation characterize recombi-
nant hirudin genetically fused to albumin. Blood.
1997;89:3243-3252.
19. Arndt KM, Muller KM, Pluckthun A. Factors inu-
encing the dimer to monomer transition of an anti-
body single-chain Fv fragment. Biochemistry.
1998;37:12918-12926.
20. Haspot F, Villemain F, Laamme G, et al. Differ-
ential effect of CD28 versus B7 blockade on di-
rect pathway of allorecognition and self-restricted
responses. Blood. 2002;99:2228-2234.
21. Perrin PJ, June CH, Maldonado JH, Ratts RB,
Racke MK. Blockade of CD28 during in vitro acti-
vation of encephalitogenic T cells or after disease
onset ameliorates experimental autoimmune en-
cephalomyelitis. J Immunol. 1999;163:1704-
1710.
22. Silver PB, Hathcock KS, Chan CC, Wiggert B,
Caspi RR. Blockade of costimulation through B7/
CD28 inhibits experimental autoimmune uveoreti-
nitis, but does not induce long-term tolerance.
J Immunol. 2000;165:5041-5047.
23. Yu XZ, Bidwell SJ, Martin PJ, Anasetti C. CD28-
specic antibody prevents graft-versus-host dis-
ease in mice. J Immunol. 2000;164:4564-4568.
24. Dong VM, Yuan X, Coito AJ, WaagaAM, Sayegh
MH, Chandraker A. Mechanisms of targeting
CD28 by a signaling monoclonal antibody in
acute and chronic allograft rejection. Transplanta-
tion. 2002;73:1310-1317.
25. Shahinian A, Pfeffer K, Lee KP, et al. Differential T
cell costimulatory requirements in CD28-decient
mice. Science. 1993;261:609-612.
26. Bachmaier K, Pummerer C, Shahinian A, et al.
Induction of autoimmunity in the absence of
CD28 costimulation. J Immunol. 1996;157:1752-
1757.
27. Wu Y, Zhou Q, Zheng P, Liu Y. CD28-independent
induction of T helper cells and immunoglobulin
class switches requires costimulation by the heat-
stable antigen. J Exp Med. 1998;187:1151-1156.
28. Kirchhoff S, Muller WW, Li-Weber M, Krammer
PH. Up-regulation of c-FLIPshort and reduction of
activation-induced cell death in CD28-costimu-
lated human T cells. Eur J Immunol. 2000;30:
2765-2774.
29. Boonen GJ, van Dijk AM, Verdonck LF, van Lier
RA, Rijksen G, Medema RH. CD28 induces cell
cycle progression by IL-2-independent down-
regulation of p27kip1 expression in human pe-
ripheral T lymphocytes. Eur J Immunol. 1999;29:
789-798.
30. Blair PJ, Riley JL, Levine BL, et al. CTLA-4 liga-
tion delivers a unique signal to resting human
CD4 T cells that inhibits interleukin-2 secretion
but allows Bcl-X(L) induction. J Immunol. 1998;
160:12-15.
31. Scheipers P, Reiser H. Fas-independent death of
activated CD4() T lymphocytes induced by
CTLA-4 crosslinking. Proc Natl Acad Sci U S A.
1998;95:10083-10088.
32. Broeren C, Gray G, Carreno B, June C. Costimu-
lation light: activation of CD4T cells with CD80
or CD86 rather than anti-CD28 leads to a Th2
cytokine prole. J Immunol. 2000;165:6908-6914.
33. Salazar-Fontana LI, Sanz E, Merida I, et al. Cell
surface CD28 levels dene four CD4T cell sub-
sets: abnormal expression in rheumatoid arthritis.
Clin Immunol. 2001;99:253-265.
34. Ferkol T, Eckman E, Swaidani S, Silski C, Davis
P. Transport of bifunctional proteins across respi-
ratory epithelial cells via the polymeric immuno-
globulin receptor. Am J Respir Crit Care Med.
2000;161:944-951.
35. Weigand K, Russi E, von Schulthess G, Bavaud
C. Turnover of autologous alpha 1-antitrypsin in a
patient with congenital analbuminemia. Klin
Wochenschr. 1983;61:547-552.
36. Weber W, Steube K, Gross V, et al. Unglycosy-
lated rat alpha 1-proteinase inhibitor has a six-
fold shorter plasma half-life than the mature gly-
coprotein. Biochem Biophys Res Commun. 1985;
126:630-635.
570 VANHOVE et al BLOOD, 15 JULY 2003 VOLUME 102, NUMBER 2
For personal use only. by guest on June 12, 2013. bloodjournal.hematologylibrary.orgFrom
... Finally, a pegylated F(ab)-fragment directed against human TNF-α made its way to the clinics for treatment of rheumatoid arthritis and Crohn's disease (Choy et al. 2002;Sandborn et al. 2007). Another option for an increase in half life would be the generation of a fusion protein containing F(ab)86 and a bigger but rather inert fusion partner like albumin (Smith et al. 2001) or α1-antitrypsin (Vanhove et al. 2003). The latter has been pursued for an Fv-single chain directed against human CD28. ...
... The latter has been pursued for an Fv-single chain directed against human CD28. Functionality of the Fv-fragment was given and half life could be extended to a useful amount of time (Vanhove et al. 2003). Taken together, F(ab)86 is a potent tool to study CD40 blockade in murine models for tolerance induction and autoimmune diseases. ...
... To prevent this, monovalent molecules against CD28 are now developed. A single chain Fv construct with α1-antitrypsin directed against CD28 has been shown to prevent allospecific T cell activation in vitro (Vanhove et al. 2003) and to prevent renal and heart allograft rejection in combination with a calcineurin inhibitor in non-human primates (Poirier et al. 2010). Together with blockade of the CD40 pathway however, CTLA-4-Ig is able to induce long term allograft acceptance in a heart allograft model in mouse , emphasizing the importance of these two costimulation pathways for allospecific immune responses. ...
Article
Kidney transplantation is the best therapy option for patients with end stage renal disease. But patients receiving a graft from a genetically non-identical donor are at a constant risk of rejection. The actual treatment for an organ recipient therefore aims to induce a systemic immunosuppression, which is very potent in inhibition of acute rejections. The long-term outcome of renal transplants, however, is still hampered by the toxicities of these regimens and their disability to prevent chronic allograft rejection. Furthermore, the systemic immunosuppression leads to a higher risk for infections and/or cancer. Therefore, new strategies for more specifc and less toxic treatments for recipients of renal grafts are needed. A major goal of research is the induction of donor-specifc tolerance. A promising tool to achieve both is costimulation blockade. Costimulation in addition to recognition of the cognate antigen is needed to fully activate a T cell. A variety of costimulatory pathways are described and their blockade has been shown to be beneficial in models for solid organ transplantation. Costimulation is also important during the interaction of a T cell with its target. Renal tubular epithelial cells (rTECs), which are the major target of cytotoxic T lymphocytes during kidney allograft rejection, do also express costimulatory molecules under inflammatory conditions. One of the most important costimulatory pathways in both processes is the CD40-CD154 pathway. A more recently described costimulatory molecule expressed on T cells is DNAM-1. In section 1 we evaluate the role of CD40 expressed on donor cells for the direct alloresponse during the priming and the effector phase in vitro. We show that donor CD40 is important for induction of allospecific Th1, Th17 and cytotoxic T cells. In an in vivo model for renal allograft rejection we show that renal allografts from CD40 deficient donors are partially protected from the effects of acute rejection. Studies blocking the CD40- CD154 pathway so far mostly used blocking antibodies against CD154. The translation of such an agent to the clinics has resulted in thromboembolic events in patients and is thus not pursued anymore. In mice the blockade of CD40 itself however is not possible with the antibodies available today. In section 2 we therefore present the development of an F(ab)-fragment against murine CD40 with exclusively antagonistic properties. F(ab)86 is able to prevent CD40 triggered B cell activation and proliferation and reduces allospecific T cell proliferation in vitro. DNAM-1 is a costimulatory molecule expressed on T and NK cells. Its two ligands CD155 and CD112 are expressed on several cell types, also in the kidney. In section 3 we show that DNAM-1 ligands are expressed on rTECs. We detected a role for DNAM-1 during allospecific T cell priming but not during cytotoxic activity against rTECs. Furthermore, we could exclude that ligation of CD155 and CD112 enhances T cell activity during allospecific priming or cytotoxicity against rTECs. The in vitro results are further strengthened by the finding that renal allografts deficient in CD155 and CD112 are not protected from rejection. Summing up, this study gives a deeper insight into the role and relevance of the CD40 and DNAM-1 pathways during renal allograft rejection and suggests a new tool to block murine CD40. Transplantation ist die beste Therapie für Patienten mit Nierenversagen. Aber Patienten die ein Transplantat von einem genetisch unterschiedlichen Spender erhalten tragen ein dauerhaftes Risiko, ihr Transplantat abzustossen. Die momentane Strategie, um das zu verhindern, ist eine systemische Immunsuppression, welche akute Abstossungen erfolgreich zu verhindern vermag. Die verwendeten Medikamente besitzen jedoch toxische Nebenwirkungen und sind nicht in der Lage chronische Abstossungen zu verhindern. Weiterhin tragen diese Patienten aufgrund der Immunsuppression ein erhöhtes Risiko für Infektionen und/oder Krebserkrankungen. Es ist also notwendig, gezieltere und weniger toxische Therapieoptionen zu finden. Eine weitere Option ist die Induktion von immunologischer Toleranz spezifisch für das transplantierte Organ. Eine mögliche Strategie, um beide Ziele zu erreichen, ist die Costimulations-Blockade. T Zellen benötigen für eine vollständige Aktivierung nicht nur ein Signal durch den T-Zell-Rezeptor, sondern zusätzlich costimulatorische Signale. Mehrere costimulatorische Signalwege sind inzwischen beschrieben. Sie zu blockieren hat sich in mehreren Modellen für Organtransplantation als förderlich erwiesen. Doch auch für die Interaktion zwischen T Zelle und Zielzelle spielt Costimulation eine Rolle. Renale Tubulus- Epithelzellen (rTECs) sind Zielzellen für allospezifische zytotoxische T Zellen während der Abstossng eines Nierentransplantats. Sie exprimieren costimulatorische Moleküle unter inflammatorischen Bedingungen. Ein wichtiger costimulatorischer Signalweg für beide erwähnten Vorgänge ist der CD40-CD154-Signalweg. Weiterhin wurde DNAM-1 als costimulatorisches Molekül auf T Zellen identifiziert. In Abschnitt 1 untersuchen wir die Rolle von CD40 auf Spender-Zellen während der T Zell Aktivierungs- und Effektorpahse in vitro. Wir zeigen, dass Donor-CD40 eine Rolle für die Induktion von Th1, Th17 und zytotoxischen T Zellen spielt. In einem in vivo-Modell für Nierentransplantation fanden wir weiterhin, dass Transplantate von CD40-defizienten Spendern trotz akuter Abstossung eine bessere Funktion aufweisen. Die meisten Studien, in denen der CD40-Signalweg bisher blockiert wurde, basierten auf der Nutzung eines Antikörpers gegen CD154. Als ein solcher Antikörper zum ersten Mal in Patienten verwendet wurde, traten jedoch thromboembolische Komplikationen auf, die eine solche Therapie unmöglich machen. In Mausmodellen ist es bisher nicht möglich gewesen CD40 selbst zu blockieren, da alle verfügbaren Antikörper agonistisch wirken. In Abschnitt 2 zeigen wir daher die Entwicklung eines F(ab)-Fragments gegen Maus-CD40, das ausschliesslich antagonistische Eigenschaften besitzt. F(ab)86 war in vitro in der Lage CD40-mediierte B Zell Aktivierung und allospezifische T Zell-Proliferation zu supprimieren. DNAM-1 ist ein costimulatorisches Molekül, das auf T und NK Zellen exprimiert wird. Seine zwei Liganden CD155 und CD112 werden auf verschiedenen Zelltypen exprimiert, unter anderem in der Niere. In Abschnitt 3 zeigen wir, dass CD155 und CD112 auf rTECs exprimiert werden. Wir konnten weiterhin feststellen, dass DNAM-1 eine Rolle während der allospezifischen T Zell Aktivierung spielt. Für die Zytotoxizität gegen rTECs hingegen wird DNAM-1 nicht benötigt. Letztlich konnten wir ausschliessen, dass die beiden Liganden CD155 und CD112 einen Einfluss auf allospezifische Aktivierung oder Zytotoxizität gerichtet gegen rTECs haben. Diese in vitro Ergebnisse konnten wir in einem in vivo-Modell erhärten. Nierentransplantate von CD155- oder CD112-defizienten Spendern sind nicht vor der Abstossung geschützt. Zusammenfassend können wir mit dieser Arbeit die Rolle von CD40 und DNAM- 1 während der Abstossung von Nierentransplantaten genauer erkärenl und eine Strategie präsentieren, um murines CD40 zu blockieren.
... We next determined whether the blockade of CD28:B7 interactions inhibits membrane transfer from Raji APCs using EM. To block B7 without activating CD28 due to crosslinking, we produced recombinant single-chain variable fragment (scFv) of a B7-blocking anti-CD28 antibody CD28.3 (Vanhove et al., 2003), abbreviated as 28.3scFv hereafter (Fig. 1 C). As a proof-of-principle, when we conjugated CD28 −/− CD28-mCherry + Jurkat T cells with CD80 −/− CD86 −/− CD80-GFP + or CD80 −/− CD86 −/− CD86-GFP + Raji APCs, 28.3scFv treatment prevented the coenrichment of CD28 and CD80/CD86 to the Jurkat:Raji interface ( Fig. 1 D). ...
Article
Full-text available
CD28 and CTLA4 are T cell coreceptors that competitively engage B7 ligands CD80 and CD86 to control adaptive immune responses. While the role of CTLA4 in restraining CD28 costimulatory signaling is well-established, the mechanism has remained unclear. Here, we report that human T cells acquire antigen-presenting-cell (APC)–derived B7 ligands and major histocompatibility complex (MHC) via trogocytosis through CD28:B7 binding. Acquired MHC and B7 enabled T cells to autostimulate, and this process was limited cell-intrinsically by CTLA4, which depletes B7 ligands trogocytosed or endogenously expressed by T cells through cis-endocytosis. Extending this model to the previously proposed extrinsic function of CTLA4 in human regulatory T cells (Treg), we show that blockade of either CD28 or CTLA4 attenuates Treg-mediated depletion of APC B7, indicating that trogocytosis and CTLA4-mediated cis-endocytosis work together to deplete B7 from APCs. Our study establishes CTLA4 as a cell-intrinsic molecular sink that limits B7 availability on the surface of T cells, with implications for CTLA4-targeted therapy.
... It has been suggested that an antagonistic CD28 antibody, which blocks CD28 costimulation but leaves CTLA-4 mediated responses intact, may be superior to CTLA-4-Ig molecules 68,73 . Indeed, it was postulated that preserved CTLA-4 signalling after treatment with antagonistic CD28 antibodies upregulated the inhibitory SLAM family member 2B4 (CD244), which then played a critical role in the observed immunoregulation 68 ; however, 2B4 upregulation was not observed in a subsequent study in non-human primates 72 . ...
Article
Full-text available
Therapeutic targeting of immune checkpoints has garnered significant attention in the area of cancer immunotherapy, in which efforts have focused in particular on cytotoxic T lymphocyte antigen 4 (CTLA4) and PD1, both of which are members of the CD28 family. In autoimmunity, these same pathways can be targeted to opposite effect: to curb the over-exuberant immune response. The CTLA4 checkpoint serves as an exemplar, whereby CTLA4 activity is blocked by antibodies in cancer immunotherapy and augmented by the provision of soluble CTLA4 in autoimmunity. Here, we review the targeting of co-stimulatory molecules in autoimmune diseases, focusing in particular on agents directed at members of the CD28 or tumour necrosis factor receptor families. We present the state of the art in co-stimulatory blockade approaches, including rational combinations of immune inhibitory agents, and discuss the future opportunities and challenges in this field.
Article
Immunosuppressive agents have enabled the development of allogenic transplantation during the last 40 years, allowing considerable improvement in graft survival. However, several issues remain such as the nephrotoxicity of calcineurin inhibitors, the cornerstone of immunosuppressive regimens and/or the higher risk of opportunistic infections and cancers. Most immunosuppressive agents target T cell activation and may not be efficient enough to prevent allo-immunization in the long term. Finally, antibody mediated rejection due to donor specific antibodies strongly affects allograft survival. Many drugs have been tested in the last decades, but very few have come to clinical use. The most recent one is CTLA4-Ig (belatacept), a costimulation blockade molecule that targets the second signal of T cell activation and is associated with a better long term kidney function than calcineurin inhibitors, despite an increased risk of acute cellular rejection. The research of new maintenance long-term immunosuppressive agents focuses on costimulation blockade. Agents inhibiting CD40-CD40 ligand interaction may enable a good control of both T cells and B cells responses. Anti-CD28 antibodies may promote regulatory T cells. Agents targeting this costimulation pathways are currently evaluated in clinical trials. Immunosuppressive agents for ABMR treatment are scarce since anti-CD20 agent rituximab and proteasome inhibitor bortezomib have failed to demonstrate an interest in ABMR. New drugs focusing on antibodies removal (imlifidase), B cell and plasmablasts (anti-IL-6/IL-6R, anti-CD38…) and complement inhibition are in the pipeline, with the challenge of their evaluation in such a heterogeneous pathology.
Article
Résumé Le développement d’agents immunosuppresseurs a permis l’essor de la transplantation rénale depuis 50 ans. Les inhibiteurs de calcineurine, à la base des stratégies immunosuppressives actuelles, sont associés à une néphrotoxicité importante. Ces traitements ne sont pas spécifiques de la réponse allo-immune et sont associés à un risque majoré d’infections et de cancers. Enfin, les traitements utilisés ciblent principalement les lymphocytes T et ne préviennent pas suffisamment le développement des anticorps anti-HLA, associés aux rejets médiés par les anticorps et cause majeure de perte des transplants au long cours. Le développement de nouveau traitements immunosuppresseurs reste donc un enjeu majeur en transplantation. Une voie prometteuse de développement de nouveaux immunosuppresseurs est le blocage de la costimulation (activation lymphocytaire). Les agents inhibant l’interaction CD40-CD40 ligands permettraient de contrôler à la fois la réponse allogénique T et B. Les anticorps anti-CD28 permettraient le développement de cellules T régulatrices. Plusieurs essais cliniques évaluant ces différents agents sont en cours. Les traitements validés en cas de rejet médié par les anticorps sont peu nombreux (échanges plasmatiques, immunoglobulines intraveineuses). Les traitements en cours de développement reposent sur l'élimination des anticorps (Imlifidase), l’inhibition de l’activation des lymphocytes B et des plasmocytes (anti-IL-6/IL-6R, anti-CD38…), l’inhibition du complément. Leur validation est rendue difficile par l’hétérogénéité des phénotypes de ce type de rejets. Enfin, des stratégies thérapeutiques innovantes afin d’obtenir une tolérance du transplant sont en cours de développement, basées sur la thérapie cellulaire (greffe de cellules souches, cellules mésenchymateuses, cellules régulatrices).
Chapter
T cells play a pivotal role in orchestrating immune responses directed against a foreign (allogeneic) graft. For T cells to become fully activated, the T-cell receptor (TCR) must interact with the major histocompatibility complex (MHC) plus peptide complex on antigen-presenting cells (APCs), followed by a second “positive” costimulatory signal. In the absence of this second signal, T cells become anergic or undergo deletion. By blocking positive costimulatory signaling, T-cell allo-responses can be aborted, thus preventing graft rejection and promoting long-term allograft survival and possibly tolerance (Alegre ML, Najafian N, Curr Mol Med 6:843–857, 2006; Li XC, Rothstein DM, Sayegh MH, Immunol Rev 229:271–293, 2009). In addition, costimulatory molecules can provide negative “coinhibitory” signals that inhibit T-cell activation and terminate immune responses; strategies to promote these pathways can also lead to graft tolerance (Boenisch O, Sayegh MH, Najafian N, Curr Opin Organ Transplant 13:373–378, 2008). However, T-cell costimulation involves an incredibly complex array of interactions that may act simultaneously or at different times in the immune response and whose relative importance varies depending on the different T-cell subsets and activation status. In transplantation, the presence of foreign alloantigen incites not only destructive T effector cells but also protective regulatory T cells, the balance of which ultimately determines the fate of the allograft (Lechler RI, Garden OA, Turka LA, Nat Rev Immunol 3:147–158, 2003). Since the processes of alloantigen-specific rejection and regulation both require activation of T cells, costimulatory interactions may have opposing or synergistic roles depending on the cell being targeted. Such complexities present both challenges and opportunities in targeting T-cell costimulatory pathways for therapeutic purposes. In this chapter, we summarize our current knowledge of the various costimulatory pathways in transplantation and review the current state and challenges of harnessing these pathways to promote graft tolerance (summarized in Table 10.1).
Article
The promising outcomes of immune‐checkpoint based immunotherapies in cancer have provided a proportional perspective ahead of exploiting similar approaches in allotransplantation. Belatacept (CTLA‐4‐Ig) is an example of costimulation blockers successfully exploited in renal transplantation. Due to the wide range of regulatory molecules characterized in the past decades, some of these molecules might be candidates as immunomodulators in the case of tolerance induction in transplantation. Although there are numerous attempts on the apprehension of the effects of co‐signaling molecules on immune response, the necessity for a better understanding is evident. By increasing the knowledge on the biology of co‐signaling pathways, some pitfalls are recognized and improved approaches are proposed. The blockage of CD80/CD28 axis is an instance of evolution toward more efficacy. It is now evident that anti‐CD28 antibodies are more effective than CD80 blockers in animal models of transplantation. Other co‐signaling axes such as PD‐1/PD‐L1, CD40/CD154, 2B4/CD48, and others discussed in the present review are examples of critical immunomodulatory molecules in allogeneic transplantation. We review here the outcomes of recent experiences with co‐signaling molecules in preclinical studies of solid organ transplantation. T cell co‐signaling molecules pose an exclusive opportunity for management of destructive immune responses during allogeneic transplantation. The manipulation in specific arms of the immune system is the hallmark of interventions in co‐signaling molecules in transplantation.
Article
Nephrotoxicity of calcineurin inhibitors and uncontrolled effector function of alloreactive T lymphocytes are main drivers of transplant dysfunctions. T lymphocytes either directly damage tissues or indirectly promote inflammation and antibody responses. Beside inhibitors of calcium-dependent pathways and antimetabolites, modulators of T-cell costimulation are elected pharmacological tools to enable interference with immune-mediated transplant dysfunctions. CD28 and CTLA-4 are major costimulatory and coinhibitory cell surface signaling molecules interacting with CD80/86, known to be critically important for immune response of committed T cells and regulation. Initial bench to beside translation, 2 decades ago, resulted in the development of belatacept CTLA-4 fused with an immunoglobulin Fc domain, a biologic inhibiting interaction of both CD28 and CTLA-4 with CD80/86. Despite proven effectiveness in inhibiting alloimmune responses, clinical use of belatacept in kidney transplantation revealed a substantially high incidence of acute, cell-mediated rejection. The cause of belatacept-resistant graft rejection was allocated to elevated pretransplant frequencies of CD28 memory T cells. Owing to different requirements in CD28 costimulatory and CTLA-4 coinhibitory signals to control naive and memory T cells, selective antagonists of CD28-CD80/86 interactions have been developed on the rationale that preservation of CTLA-4-mediated regulatory mechanisms would result in a better control of alloreactivity and would represent a regulatory T-cell-compatible immunosuppression. After the successful testing of selective CD28 antagonists in First In Human studies, this review delineates how this shift in paradigm performed in preclinical transplantation models and evaluates its clinical potential.
Article
Selection of a suitable assay to measure the activity of drugs based on released-active forms (RA forms of Abs) is an important step during their investigation. In this study, ELISA was utilized to examine the effect of RA forms of Abs to interferon gamma on the interaction between monoclonal anti-interferon gamma antibodies and human interferon gamma. The data showed that such RA forms of Abs are able to modulate the antibody interaction with interferon gamma, and it was suggested that the observed influence of RA forms of Abs on ‘antibody-antigen’ interaction could be used to analyze the activity of this kind of drugs.
Article
Full-text available
We have assessed the functional effects of a panel of CTLA-4 mAbs on resting human CD4+ T cells. Our results demonstrate that some CTLA-4 mAbs can inhibit proliferative responses of resting CD4+ cells and cell cycle transition from G0 to G1. The inhibitory effects of CTLA-4 were evident within 4 h, at a time when cell surface CTLA-4 expression remained undetectable. Other CTLA-4 mAbs had no detectable inhibitory effects, indicating that binding of Ab to CTLA-4 alone is not sufficient to mediate down-regulation of T cell responses. Interestingly, while IL-2 production was shut off, inhibitory anti-CTLA-4 mAbs permitted induction and expression of the cell survival gene bcl-X(L). Consistent with this observation, cells remained viable and apoptosis was not detected after CTLA-4 ligation.
Article
Full-text available
It is well established that B7-CD28/CTLA4 interactions play an important role in the induction of T helper cells for T-dependent antibody responses. However, targeted mutation of CD28 does not significantly affect production of IgG and activation of CD4 T helper cells in response to infections by some viruses and nematode parasites. To test whether the CD28-independent induction of Ig class switches requires costimulation by the heat-stable antigen (HSA), we compared T helper cell induction and antibody response in mice deficient for either HSA, CD28, or both genes. We found that after immunization with KLH-DNP, mice deficient for both CD28 and HSA lack DNP-specific IgA and all subtypes of IgG. This deficiency corresponds to a reduced number of effector helper T cells that rapidly produce IL-2, IL-4, and IFN-γ after in vitro stimulation with carrier antigen KLH. In contrast, priming of T helper cells and Ig class switch are normal in mice deficient with either HSA or CD28 alone. IgM responses are not affected by any of these targeted mutations. These results demonstrate that CD28-independent induction of T helper cells and Ig class-switches requires costimulation by the HSA.
Article
CD28 is the primary T cell costimulatory receptor, and upon ligation with its ligands, it enhances T cell proliferation and IL-2 synthesis. In this study we examined the role of CD28 in the initial proliferative response and cell cycle entry of T lymphocytes. Stimulation through CD3 alone resulted in a poor proliferative response, while in the presence of CD28 costimulation a strong increase in the number of cells in S-phase could be detected after 48 h of stimulation. CD28 costimulation enhanced expression of cyclin D3 and induced down-regulation of p27kip1 expression. Cross-linking CD28 was much more effective in inducing cyclin D3 expression and in down-regulating p27kip1 expression than addition of IL-2. Blocking experiments, using antibodies that neutralize IL-2 or the IL-2 receptor, showed that the effects induced by CD28 are independent of endogenous IL-2. Moreover, using a variety of immunosuppressants that interfere with IL-2 signaling pathways, we were able to show that IL-2 is not required for cell cycle entry induced by CD28 costimulation. From these experiments it can be concluded that CD28 and IL-2 use different signaling pathways for down-regulation of p27kip1 expression. We hypothesize that costimulation through CD28 is responsible for initial cell cycle entry of T lymphocytes, while IL-2, which is produced after costimulation, might be involved in sustaining proliferation.
Article
The carbohydrate structure Gal alpha1,3Gal expressed on pig cells is the major antigen recognized by xenoreactive natural antibodies in the higher primates. In xenotransplantation, natural antibodies binding to that structure initiate hyperacute rejection, and the anti-Gal alpha1,3Gal antibodies that are elicited probably take part in later phases of vascularized graft rejection. This epitope also appears to be involved in innate cellular responses. Inactivation of alpha1,3 galactosyltransferase in transgenic pigs would certainly lead to the success of xenotransplantation, but gene knockout in pigs is not feasible yet. As a novel strategy to inhibit alpha1,3 galactosylation, we generated recombinant single-chain Fv (ScFv) antibodies directed against pig alpha1,3-galactosyltransferase and evaluated the effect of their intracellular expression on enzyme activity and Gal alpha1,3Gal expression. After in vitro transfection in pig cells, the scFv antibody anti-pig alpha1,3-galactosyltransferase reduced the amount or function of enzyme by up to 70% as evidenced by immunofluorescence and measurement of cell-associated activity. Consequently, Gal alpha1,3Gal on cell membranes was reduced to the same extent. This led to a profound (more than 90%) reduction in the cytotoxicity involving anti-Gal antibodies and complement. Although not sufficient to knock out the overall human anti-pig natural xenoreactivity, intracellular expression of the scFv antibody anti-alpha1,3-galactosyltransferase in pig cells significantly decreases the amount of Gal alpha1,3Gal and could be important to protect cells from elicited antibodies as well as from innate effectors.
Article
Films of 1,3,4,6-tetrachloro-3a,6a-diphenylglycoluril (conveniently “plated” in reaction vessels from methylene chloride solution) react rapidly in the solid phase with aqueous mixtures of I− and proteins to yield iodinated proteins. Similarly applied, this reagent brings about iodination of cell membranes, although, apparently, at a somewhat lesser rate than in iodinations carried out with lactoperoxidase-glucose oxidase. The stability and sparing solubility of this chloroglycoluril in water can account for the minimal damage to proteins and living cells observed in these iodinations; further, these properties allow for elimination of the reduction step employed at the close of iodinations with soluble chloroamides such as chloramine-T.
Article
Films of 1,3,4,6-tetrachloro-3a,6a-diphenylglycoluril (conveniently “plated” in reaction vessels from methylene chloride solution) react rapidly in the solid phase with aqueous mixtures of I− and proteins to yield iodinated proteins. Similarly applied, this reagent brings about iodination of cell membranes, although, apparently, at a somewhat lesser rate than in iodinations carried out with lactoperoxidase-glucose oxidase. The stability and sparing solubility of this chloroglycoluril in water can account for the minimal damage to proteins and living cells observed in these iodinations; further, these properties allow for elimination of the reduction step employed at the close of iodinations with soluble chloroamides such as chloramine-T.
Article
The binding of antigen to the multicomponent T cell antigen receptor (TcR)-CD3 complex leads to the activation of several signal transduction pathways which result in T lymphocyte proliferation and lymphokine secretion by molecular mechanisms and catalytic molecules as yet poorly defined. One of the earliest events that follows the triggering of the antigen-specific TcR-CD3 complex is a rapid tyrosine phosphorylation of several intracellular substrates, suggesting stimulation of at least one protein tyrosine kinase (PTK). Since none of the seven TcR-CD3 subunits exhibits a recognizable kinase domain, it seems likely that the receptor complex is associated with an intracellular PTK. p59fyn and the T lymphocyte-specific p56lck are two intracellular, non-receptor, cell membrane-associated PTK of the src family expressed in T lymphocytes. Here, we show by double immunofluorescence microscopy a specific co-distribution of p59fyn, but not p56lck, with antibody-induced TcR or CD3 caps in intact human T lymphocytes. These findings provide direct evidence for a significant association of p59fyn with the TcR-CD3 complex under physiologically relevant conditions in functional T lymphocytes. They suggest that p59fyn is a crucial component of the TcR signal transduction machinery and that one of the earliest consequences of antigen recognition by the TcR is p59fyn-mediated phosphorylation of intracellular substrates on tyrosine residues.
Article
The murine monoclonal antibody A7 (Mab A7) against human colon cancer was chemically modified with methoxypolyethylene glycol (PEG) (Mr 5000). A high substitution of PEG molecules on Mab A7 produced a progressive reduction in antibody-binding activity. The pharmacokinetic and immunological properties of PEG-modified monoclonal antibody A7 (Mab A7) and the PEG-modified F(ab')2 fragment, which retained their antibody-binding activity, were assessed and compared with the parent Mab A7 and the parent F(ab')2 fragment. Blood clearance of PEG-modified antibodies appeared to be diminished by PEG modification and was fitted by a two-compartment model. Low PEG-substituted Mab A7 showed less organ uptake in the liver and spleen and similar uptake in the lung and kidney, compared with the parent Mab A7. PEG-F(ab')2 showed less uptake in the liver and kidney. Both preparations exhibited less tissue:blood ratios in all resected organs as compared with parent antibodies. Tumor localization was enhanced by PEG modification for the F(ab')2 fragment, but not by PEG modification for the whole Mab A7. Multiple i.v. administration of PEG-modified antibody to rabbit did not appear to elicit a measurable immune response to the antibody portion of the conjugate. In conclusion, PEG-modified antibodies are promising reagents as drug carriers to the target tumor.