Sei sulla pagina 1di 10

REVIEWS

B cells and transplantation tolerance


Allan D. Kirk, Nicole A. Turgeon and Neal N. Iwakoshi
Abstract | Transplantation tolerance is a state of immune unresponsiveness (or benign responsiveness) to the
presence of specific, nonself antigens in the absence of chronic immunosuppressive therapy. Renal transplant
tolerance remains a desired yet generally unattained goal that would enable transplantation to be performed
without the risk of graft rejection or the need for broadly immunosuppressive drugs, which can have toxic
effects. Studies published in the past few years have provided evidence that B cells have an important role
in both graft rejection and transplantation tolerance. Indeed, antibody-dependent and antibody-independent
functions of B cells account for both tolerogenic and rejection-promoting immune responses in transplant
recipients. This Review comprises a discussion of the mechanisms involved in the induction of B-cell tolerance
and a survey of current and emerging therapies that target the effects of B cells in transplant recipients.
Kirk, A. D. et al. Nat. Rev. Nephrol. 6, 584–593 (2010); published online 24 August 2010; doi:10.1038/nrneph.2010.111

Introduction Physiological self tolerance


In the past, T‑cell‑mediated mechanisms were con‑ B‑cell tolerance of self antigens is established through
sidered the main cause of renal allograft rejection. multiple mechanisms that take place at various stages of
Although improvements in T‑cell‑directed immuno‑ B‑cell development and differentiation (Figure 1). These
suppression have decreased the incidence of acute processes are only briefly outlined in this article as they
cellular rejection,1,2 humoral immune responses, specifi‑ have been thoroughly discussed elsewhere.12,13
cally those mediated by alloantibodies (that is, by anti‑ Much of our knowledge of these mechanisms has
bodies produced by the graft recipients that react with been derived from studies of autoreactive B cells. B‑cell
graft isoantigens), have increasingly been recognized receptors are assembled through a stochastic process of
as causes of renal allograft rejection. Indeed, humoral V(D)J genetic recombination that takes place in devel‑
immune responses can lead to renal allograft rejection oping B‑cell precursors. The membrane‑bound anti‑
even in patients whose cell‑mediated immune responses body moiety of the resulting B‑cell receptors comprises
are well controlled. a large repertoire of randomly selected heavy and light
The overall incidence of antibody‑mediated rejec‑ chains. The receptor diversity achieved via this approach
tion (AMR) is estimated to be 2–10% for kidney trans‑ is enormous, and a substantial number of receptors with
plant recipients.3,4 The frequency of AMR is increased autoreactive potential are produced. Indeed, 20–50% of
in individuals with a previously failed allograft and in B‑cell receptors generated by V(D)J genetic recombina‑
sensi tized patients (that is, those who have allo‑ tion are thought to carry an unacceptably high affinity
antibodies before renal transplantation as a result of for self antigens. Consequently, B‑cell maturation incor‑
pregnancy, blood transfusions or prior transplantation porates processes that reduce the proportion of auto‑
procedures). AMR presents as graft dysfunction in the reactive B cells, thereby helping to prevent autoimmune
presence of donor‑specific antibodies, and its presence diseases.14–17 In general, these processes belong to two
is indicated in biopsy samples by evidence of comple‑ categories: those that eliminate autoreactive B cells in
ment deposition (typically deposition of C4d).5–7 The the central and peripheral lymphoid organs, and those
development of donor‑specific antibodies and histo‑ that prevent activation and differentiation of surviv‑
pathology consistent with AMR correlates with poor ing autoreactive B cells (which are generally not as
long‑term allograft survival. In this Review, we provide strongly autoreactive as the B cells eliminated in the
an overview of the physiological mechanisms of self tol‑ lymphoid organs) into plasma cells during an immune
Emory Transplant erance and describe the currently available and emerg‑ response. Although the specific mechanisms that deter‑
Center, Department of
Surgery, Emory ing therapies that target B cells in the setting of renal mine which of these pathways are induced in a par‑
University, transplantation. A complete discussion of the mecha‑ ticular autoreactive B cell are unclear, elimination of
101 Woodruff Circle,
WMB 5105 Atlanta,
nisms underlying AMR is beyond the scope of this high‑affinity autoreactive receptors tends to involve
GA 30322, USA Review, but these mechanisms have been extensively clonal deletion and receptor editing, whereas elimina‑
(A. D. Kirk, reviewed elsewhere.8–11 tion of low‑affinity autoreactivity tends to involve
N. A. Turgeon,
N. N. Iwakoshi). anergy. As a result of these processes, less than 10%
of the immature B cells formed in the bone marrow
Correspondence to:
A. D. Kirk Competing interests actually reach the periphery of the lymphatic system as
adkirk@emory.edu The authors declare no competing interests. transitional B cells.

584 | OCTOBER 2010 | vOlumE 6 www.nature.com/nrneph


© 2010 Macmillan Publishers Limited. All rights reserved
f o C u S o N To l E R A N C E I N T R A N S p l A N TAT I o N

Mechanisms in central lymphoid tissues Key points


Clonal deletion ■ Transplantation tolerance is currently an unattained goal; however, awareness
In the primary lymphoid organs, immature B cells that is increasing that B cells might have an important role in both graft rejection
express strongly autoreactive receptors are induced to die and tolerance
by interaction with self antigen via ‘clonal deletion’.18 If ■ In general, strategies to remove alloantibodies or suppress their production
stimulation of a B‑cell receptor by a self antigen results in have met with limited therapeutic success
intracellular signaling of sufficient strength, the immature ■ Emerging treatments for antibody-mediated renal allograft rejection need to be
B cell rapidly internalizes the autoreactive B‑cell recep‑ assessed in randomized controlled studies
tor and temporarily halts its maturation.19–21 If a B cell ■ The achievement of sustained transplantation tolerance might require induction
that possesses an autoreactive receptor fails to attenuate of B-cell tolerance to specific donor antigens
or eliminate its autoreactivity, cell death occurs within a ■ To be clinically useful, strategies for inducing B-cell tolerance must integrate
few days, either in the bone marrow or shortly after the safely into existing immunosuppressive regimens as well as modify the function
B cell arrives in the periphery of the lymphatic system and make-up of relevant B-cell subsets
as a transitional (or stage T1/T2) B cell (Figure 1).21,22
B‑cell‑receptor‑induced cell death pathways are induced
through increased levels of Bcl2‑like protein 11. This Ignorance
proapoptotic factor inhibits expression of essential B‑cell Autoreactive B cells that have a low affinity for systemic,
survival proteins of the Bcl2 family.23–25 sequestered or tissue‑specific antigens can bypass clonal
several other regulatory mechanisms associated with deletion, receptor editing and anergy and persist in the
this arrest of maturation promote self tolerance. Adhesion population of naive B cells. Collectively, these B cells are
molecules responsible for the migration of B cells, such as described as clonally ignorant and are regulated by a
l‑selectin and receptors specific for tumor necrosis factor variety of extrinsic mechanisms, as discussed below.40–42
(TnF) ligand superfamily member 13B (TnFsF13B, also
known as BAFF or Blys), a circulating cytokine required Mechanisms extrinsic to B cells
to sustain the survival of B cells in the periphery, fail to A new hypothetical mechanism has emerged in which
be induced in persistently autoreactive B cells.21,26 transitional B cells in peripheral lymphoid tissues
compete for limited availability of a follicular growth
Receptor editing factor, now identified as TnFsF13B.43–46 In contrast to
RAG1 and RAG2, the genes that encode the core clonal deletion in the bone marrow, which is intrinsically
enzymes responsible for V(D)J recombination, con‑ mediated by the B‑cell receptor, the regulation of periph‑
tinue to be expressed in autoreactive B cells that are not erally located autoreactive B cells is thought to involve a
eliminated by clonal deletion. Persistent expression of cell‑extrinsic mechanism that is responsive to homeo‑
RAG genes favors further V(D)J recombination of new stasis of the peripheral B‑cell population. Regulation
light chains. This process enables B cells that initially of peripheral B‑cell activity during the initiation of an
bear an autoreactive receptor to attenuate or overcome immune response and in germinal center reactions might
this autoreactivity.27–29 involve antigen availability, competition for develop‑
mental growth factors, T‑cell co‑stimulation and/or
Mechanisms in peripheral lymphoid tissues proinflammatory mediators.
Anergy
Autoreactive B cells that elude clonal deletion and recep‑ Current therapies for AMR
tor editing can be regulated by clonal anergy, which is Treatments that address B‑cell‑mediated mechanisms
defined as the inability of autoreactive cells to respond of graft rejection and induce alloantigen‑specific B‑cell
to stimulation by self antigen in the periphery. Anergy tolerance might enable renal allograft transplantation
is induced in transitional B cells at stage T1 and T2 to be performed in sensitized patients. These trans‑
and in mature B cells (Figure 1), and is characterized plant recipients are among the most difficult to treat.
by molecular changes that reduce the capability of Although the T‑cell alloimmune responses of sensitized
autoreactive B cells to become activated.30–36 patients can be ameliorated with current drug regimens,
A number of well‑documented intrinsic regulatory their antibody‑dependent effector responses remain
mechanisms are involved in the induction of anergy poorly controlled with therapies that target alloantibody
in B cells. Among the observed changes that involve production or induce generalized suppression of
an increase in the threshold of B‑cell activation are a humoral immunity.
modulation of the level of the IgM antibody receptor
on the surface of B cells, reduced intracellular signaling Removal of alloantibodies
resulting from B‑cell‑receptor stimulation and inhibition strategies to halt or suppress alloantibody production
of immunogenic nuclear factor κB signaling pathways. have generally met with limited therapeutic success. In
Continuous B‑cell‑receptor signaling in anergic cells patients with evidence of alloantibody sensitization before
maintains tolerogenic pathways mediated by eRK and transplantation (as determined by cellular cross‑match
nFAT signaling by mechanisms that are as yet unclear, assays and solid‑phase antibody testing) and in allograft
and induces expression of the proapoptotic protein, recipients with evidence of AMR, treatments that remove
Bcl2‑like protein 11.33,37–39 alloantibodies have resulted in fair allograft survival.3

nATuRe ReVIews | NEphRology VoluMe 6 | oCToBeR 2010 | 585


© 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

Central lymphoid tissues Peripheral lymphoid tissues


Memory
B cell
Receptor
editing
Pre-B cell T1/2 Mature B cell Differentiation

Plasma
cell
Immature,
self-reactive Self-antigen Tolerance Tolerance
B cell interaction induction induction

Induced cell death Anergic/ignorant Anergic/ignorant


B cell B cell
B-cell
tolerance Central Peripheral

Receptor editing
Clonal deletion

Anergy
Ignorance

Strategies to
■ Mixed chimerism ■ Lymphocyte depletion
induce B-cell
■ Neonatal tolerance ■ Co-stimulation blockade
tolerance
Figure 1 | Physiological mechanisms of B-cell tolerance. Immature autoreactive B cells undergo clonal deletion and
receptor editing in primary lymphoid tissues. When strongly autoreactive B cells interact with self antigens, cell death
rapidly ensues (clonal deletion). Weakly autoreactive B cells that bypass deletion will undergo receptor editing, which
results in removal or attenuation of their autoreactivity. Anergy and ignorance begin in the primary lymphoid tissues but
predominantly occur in secondary lymphoid tissues. Emerging strategies to induce B-cell tolerance in transplantation
involve neonatal tolerance, mixed chimerism, lymphocyte depletion and co-stimulation blockade. Abbreviation: T1/T2,
transitional B cell.

Plasma exchange nonselectively eliminates allo‑ bortezomib, have the potential to achieve at least partial
antibodies, but the efficacy of this strategy in improving or temporary suppression of alloantibody production
allograft survival in these patients is limited by a com‑ and/or antibody‑mediated effector functions, they do
pensatory physiological increase in antibody production, not achieve stable transplantation tolerance. Importantly,
which is induced by hypogammaglobulinemia. Infusions the safety of the broad inhibition of humoral immunity
of nonspecific immunoglobulin (IVIG) might limit this achieved by these agents cannot be assumed and requires
homeostatic antibody production. such infusions might confirmation in appropriately designed trials.
also neutralize the de novo production of donor‑specific
antibodies via anti‑idiotypic interactions between vari‑ Rituximab
able regions of IVIG and human leukocyte antigen Rituximab is a chimeric (mouse and human) CD20‑
(HlA)‑specific antibodies, inhibition of the comple‑ specific monoclonal antibody that inhibits B‑cell pro‑
ment system and saturation of the receptors involved in liferation and induces apoptosis by antibody‑dependent
antibody homeostasis, including IgA Fc receptors and and complement‑dependent cellular cytotoxicity. 48
neonatal IgA Fc receptors. Administration of rituximab results in a substantial
standard immunosuppressive agents (such as cortico‑ and rapid depletion of cells that express CD20, includ‑
steroids, azathioprine, ciclosporin, mycophenolate ing early pre‑B‑stage cells through to cells at late
mofetil, tacrolimus and sirolimus) have essentially stages of B‑cell differentiation (but excluding mature
no efficacy in treating AMR.3,47 The ineffectiveness of antibody‑secreting plasma cells).
current immunosuppressive therapies against persistent Results from small, noncontrolled studies and case
alloantibody production, along with current organ‑ reports indicate that rituximab helps to reverse AMR
allocation policies in the us, has resulted in increased when used as part of a complex multidrug treatment
numbers of alloantibody‑sensitized patients on the renal regimen. Regimens that include rituximab in combina‑
transplantation waiting list. tion with pulsed steroids, plasmapheresis, IVIG, anti‑
lymphocyte globulin and splenectomy have been studied
generalized suppression of humoral immunity in patients with AMR that is resistant to conventional
Although agents that achieve broad suppression of treatment with plasma exchange and IVIG.49–55 such
humoral immunity, such as rituximab, eculizumab or treatments have resulted in improved graft function, and

586 | OCTOBER 2010 | vOlumE 6 www.nature.com/nrneph


© 2010 Macmillan Publishers Limited. All rights reserved
f o C u S o N To l E R A N C E I N T R A N S p l A N TAT I o N

have enabled successful kidney transplantation across peritubular capillaries in renal biopsy specimens.5–7 A
ABo incompatibility barriers.49–55 However, conclusions report published in 2009 described a patient who had
on whether these positive results are the consequence severe AMR being successfully treated using eculizumab
of rituximab’s mode of action cannot be drawn since, in in combination with multiple other interventions.64
these studies, this drug was administered concomitantly Another group of investigators have presented their
with other therapies. Furthermore, CD20 is not expressed preliminary experience of eculizumab treatment at the
on the surface of mature, antibody‑secreting plasma time of kidney transplantation in a series of cross‑match‑
cells and the observed beneficial effects of rituximab positive kidney transplant recipients. Results from this
administration have been far too rapid to indicate the study showed that C5 blockade prevented the develop‑
presence of a mechanism involving decreased antibody ment of AMR in patients who developed high levels of
production or increased clearance. Thus, any therapeutic donor‑specific antibodies after transplantation.65 long‑
benefit of rituximab in patients with AMR probably term, placebo‑controlled studies are needed to evaluate
involves antigen‑mediated or cytokine‑mediated effects the efficacy of this novel approach in the prevention
on B cells. Randomized controlled trials are clearly of AMR.
necessary to understand how this and other therapies
that deplete B‑cell numbers help to prevent AMR. Emerging targets for B‑cell tolerance
B‑cell tolerance seems to be much more difficult to
Bortezomib achieve in humans than in animal models of allograft
Bortezomib is a selective inhibitor of the 26s protea‑ transplantation.66–69 This divergence probably results
some.56 In animal studies, bortezomib was effective in from species‑specific factors, different exposure to
suppressing autoantibody production by nonmalignant environmental antigens, different extents of T‑cell and
plasma cells.57,58 In these studies, normal plasma cells B‑cell diversity and different frequencies of primed
were also hypersensitive to proteasome inhibition owing alloreactive T‑cell and B‑cell precursors owing to hetero‑
to their high levels of protein biosynthesis. Proteasome logous immunity, to name a few aspects. nonetheless,
inhibition by bortezomib eliminates both short‑lived observations derived from animal models of allograft
and long‑lived plasma cells by activating the terminal transplantation have served as the foundation for modern
unfolded protein response. This response regulates clinical strategies that aim to achieve allograft tolerance
protein‑biosynthesis homeostasis within the endoplasmic by targeting B cells.
reticulum (eR); excessive eR stress leads to activation
of cell‑death pathways.59 In vitro data show a signifi‑ Neonatal tolerance
cant reduction in the production of antibodies directed Immature B cells, such as those of infants, are particu‑
against HlAs following the bortezomib‑induced apop‑ larly susceptible to the induction of tolerance. owen
tosis of plasma cells.60 In addition, results from prelimi‑ demonstrated that fraternal twin calves that had a shared
nary clinical studies suggest some efficacy of bortezomib placental circulation permanently accepted transplanted
in renal transplant recipients with AMR61 and in renal skin grafts from their twin, whereas inter‑twin skin grafts
transplant recipients with high levels of donor‑specific were rejected by calves with separate placental circula‑
antibodies before transplantation.62 However, in each of tions.70 In a subsequent study, allograft tolerance was
these studies, bortezomib was administered in combina‑ induced in mice by fetal infusion of alloantigens, which
tion with other desensitizing therapies, which limits the might have led to the clonal deletion of B cells with speci‑
conclusions that can be drawn about the utility of borte‑ ficity for these antigens.71 neonatal tolerance also occurs
zomib in treating AMR in renal transplant recipients. The naturally in human monochorionic dizygotic twins who
need to evaluate this new therapy in prospective random‑ have different blood groups; such twins become tolerant
ized and placebo‑controlled studies was highlighted by to each other’s blood group in utero.72
the results of a 2010 study, in which bortezomib alone The risk of antibody‑mediated hyperacute rejec‑
did not decrease the levels of donor‑specific antibodies tion has generally been thought to preclude ABo‑
in sensitized kidney transplant recipients.63 incompatible transplantation. However, infants do not
usually produce ABo antibodies until 5–6 months of age,
Eculizumab which represents a window of opportunity during which
unlike rituximab and bortezomib, eculizumab does not ABo‑incompatible transplantation carries a reduced
act directly on B cells, but instead suppresses antibody‑ risk of hyperacute allograft rejection.73 In 1996, a clini‑
mediated cell destruction by preventing activation of the cal trial was conducted that included 10 infant recipients
complement system. eculizumab is a humanized mono‑ of ABo‑incompatible heart grafts. standard immuno‑
clonal antibody with specificity for complement compo‑ suppression was employed, and no aggressive therapies
nent C5 (C5). Binding of eculizumab to C5 inhibits the to remove donor‑specific antibodies were implemented.74
cleavage of C5 into C5a and C5b and thereby prevents eight patients survived, and no hyperacute rejections
the formation of the membrane‑attack complex. or problems attributable to ABo incompatibility were
Although the role of the complement activation on observed. This clinical experience has subsequently
the pathogenesis of AMR is unclear, episodes of AMR been reproduced.75 The mechanism by which the ABo‑
are generally accompanied by evidence of early comple‑ incompatible heart graft is tolerated seems to involve
ment activation as demonstrated by C4d staining of the deletion of ABo‑specific B cells that persists as long as

nATuRe ReVIews | NEphRology VoluMe 6 | oCToBeR 2010 | 587


© 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

the ABo‑incompatible heart is present. Interestingly, HlA‑haploidentical individuals (that is, between a parent
this deletion seems to be transient as B cells specific for and a child) has been implemented in a pilot study in
the previously mismatched ABo epitope reappeared in patients without malignant disease. These patients
patients who subsequently underwent retransplantation received haploidentical bone marrow grafts solely to
with an ABo‑compatible heart graft.76 Thus, in contrast induce tolerance to a kidney transplant from the same
to neonatal tolerance, which seems to involve centrally donor.87 The haploidentical grafts induced transient, very
based mechanisms, allograft tolerance seems to require limited mixed chimerism that was associated with renal
peripheral antigen. allograft tolerance in four of five treated patients who
The findings of a 2004 study of the B‑cell repertoire were followed for up to 6 years.88 In the fifth patient, the
in infant recipients of ABo‑incompatible heart grafts renal allograft was lost soon after transplantation owing
suggested that B‑cell tolerance to donor‑specific blood‑ to acute AMR. Interestingly, two of the graft recipients
group antigens can develop spontaneously.77 Although who demonstrated transient mixed chimerism devel‑
the development of tolerance to donor HlAs was not oped alloantibodies and one of these individuals showed
demonstrated in these patients, tolerance to these ABo‑ clinical signs of AMR. This evidence indicates that trans‑
incompatible heart grafts provided the first demonstra‑ plantation tolerance can coexist with some level of B‑cell
tion that neonatal tolerance could be acquired in humans. reactivity to donor alloantigens. The relationship between
ABo‑incompatible transplantation has been attempted transient mixed chimerism and B‑cell‑mediated tolerance
in pediatric candidates for renal and liver transplantation to allograft transplantation has not been established.
with some success, although the opportunity for neonatal Rejection of solid‑organ grafts can still occur despite
transplantation of these organs is uncommon.52,55,78,79 the presence of mixed hematopoietic chimerism in non‑
However, in these patients, aggressive therapies were human primates, which indicates that the mechanisms
implemented to remove donor‑specific antibodies and involved in solid‑organ rejection might rely on cell types
suppress B‑cell function before and after transplantation. that persist despite mixed chimerism.89 The clinical
undoubtedly, increased understanding of the tolerogenic importance of alloantibody formation in the presence
mechanisms in infants will enable valuable insight to be of apparent T‑cell tolerance to a renal allograft is cur‑
gained into possible routes for achieving B‑cell tolerance rently unclear, but development of alloantibodies typi‑
to allografts in adult patients. cally has a negative prognostic influence in human renal
transplant recipients.90 loss of chimerism in humans
Chimerism is typically not followed by graft rejection.87 Given the
In contrast to the neonatal chimerism described by transient nature of the chimerism achieved, long‑term
owen in 1945,70 achieving chimerism in adults is chal‑ transplantation tolerance achieved in human recipients
lenging, largely owing to the immune barrier imposed by of HlA‑mismatched kidney transplants combined with
recipient T cells. This barrier can be overcome in small bone‑marrow infusion is unlikely to result from clonal
animal models as bone marrow engraftment has reli‑ deletion of B cells alone. In humans, therefore, some of
ably been shown to induce durable tolerance to the most the other mechanisms of peripheral B‑cell tolerance,
immunogenic allografts. discussed above, might be involved.
In mixed chimerism, donor and recipient hemato‑
poietic components coexist in primary lymphoid organs, lymphocyte depletion
which facilitates the clonal deletion of T cells and B cells Targeted inhibition and/or depletion of lympho‑
that react to donor‑specific alloantigens.80–83 Mixed chi‑ cytes with polyclonal or monoclonal antibodies non‑
merism, in contrast to full chimerism, can generally be specifically reduces the levels of both T‑cell and B‑cell
achieved without myeloablation and has the advantage precursors that are specific for alloantigens. Although
of preserving the graft recipient’s immunocompetence the two most common lymphocyte‑depleting agents,
while decreasing their risk of graft‑versus‑host disease. alemtuzumab and rabbit antithymocyte globulin (ATG)
However, current protocols to achieve mixed chimerism predominantly deplete T cells, both also deplete B cells.
in humans depend on a rigorous early conditioning This approach might, therefore, be a viable induction
regimen that variably includes T‑cell depletion, transient strategy for reducing alloantibody titers before trans‑
maintenance immunosuppression and thymic or total plantation or to prevent early and late graft rejection. of
lymphoid irradiation to prevent rejection of the bone‑ note, plasma cells and some B‑cell subsets are resistant
marrow graft.84,85 once established, mixed chimerism to lymphocyte‑depletion therapy, which might pose an
of multilineage hematopoietic cells is associated with important barrier to the use of these agents in inducing
lifelong tolerance of T cells and B cells (mediated by allograft tolerance (Figure 2).
clonal deletion) that enables the acceptance of any donor lymphocyte depletion might, however, decrease the
allograft without the need for immunosuppression.69 risk of allograft rejection. Furthermore, the cell popula‑
Mixed chimerism has been used clinically to achieve tions that re‑emerge after lymphocyte depletion might
transplantation tolerance to renal allografts in patients be more susceptible than the original populations to
with multiple myeloma and consequent renal failure.85,86 peripheral tolerogenic mechanisms. Consistent with
In these studies, the bone‑marrow donor was fully this hypothesis, lymphocyte depletion has enabled the
HlA‑matched to the recipient. A similar approach, reduction of maintenance immunosuppressive therapy
which involved bone‑marrow transplantation between in many allograft recipients, a phenomenon dubbed

588 | OCTOBER 2010 | vOlumE 6 www.nature.com/nrneph


© 2010 Macmillan Publishers Limited. All rights reserved
f o C u S o N To l E R A N C E I N T R A N S p l A N TAT I o N

‘prope’ (derived from the latin word for near) tolerance, 1 Abatacept, belatacept 3 Alemtuzumab
which describes incomplete donor‑specific tolerance or (anti-CD52 Ab)

52
tolerance that requires minimal immunosuppression.91

CD
However, the effects of alemtuzumab and rabbit ATG CD28 B7.1/2 6
therapies on B‑cell tolerance have not been evaluated. MCH CD20 Rituximab
CD4+ TCR (anti-CD20 Ab)
Alemtuzumab is a humanized, monoclonal antibody T cell
B cell
directed against CD52. This agent induces profound D5
2 CD154 CD40 BC
C R Alloantigen
T‑cell depletion and moderate B‑cell and monocyte
depletion. CD52 is not expressed by plasma cells or 3 2 BAFF-R
lymphocyte precursors and memory T cells and memory Alemtuzumab Anti-CD40 Ab, TACI
(anti-CD52 Ab) anti-CD154 Ab BCMA
B cells are refractory to alemtuzumab‑mediated deple‑
tion, which might limit the utility of alemtuzumab to 4 BR3-Fc
induce transplantation tolerance.92,93 BAFF blockade 5
Induction therapy with alemtuzumab in combina‑ IVIG
4
Plasmapheresis
tion with low‑dose immunosuppressive maintenance Belimumab
BAFF-specific mAb
therapy has shown promising results in terms of graft
survival in renal transplant recipients. 94–106 However, 4
despite the proven efficacy of alemtuzumab in decreas‑ Atacicept (TACI–Ig)
Plasma cell
ing the number of B cells, therapy with this monoclonal BAFF and April blockade
Figure 2 | Potential strategies for treating antibody-mediated rejection (AMR).
antibody did not prevent AMR. In fact, treatment with Dendritic
Abatacept andcell
belatacept (1) are fusion proteins that compete with CD28 for
this agent might be associated with increased rates of binding with B7.1 (CD80) or B7.2 (CD86) and block T-cell co-stimulation of B-cell
AMR, as demonstrated by C4d staining of peritubular activation and antibody production. Efforts to target the pathway associated with
capillaries, particularly when alemtuzumab is admin‑ the interaction between CD40 and CD154 (2) have been uniformly successful in
istered without a calcineurin inhibitor or to patients controlling humoral responses in animal models. Alemtuzumab (3) is a humanized
with evidence of prior subclinical sensitization. 96,107 mAb directed against CD52 that induces T-cell depletion with moderate depletion
Homeostatic lymphocyte proliferation activates T cells, of B cells and monocytes. Members of the TNF superfamily of proteins and of the
TNF superfamily of ligands regulate humoral immunity by controlling B-cell survival
and thus might promote T‑cell‑mediated B‑cell anti‑
and differentiation. Belimumab and BR3-Fc (4) are TNFSF13B-specific inhibitors.
body production. Phase II trial reports published in Atacicept (or TACI–Ig) (4) inhibits both TNFSF13B and TNFSF13. Plasmapheresis
the past 2 years or so revealed that use of alemtuzumab and IVIG (5) enable the nonselective elimination or suppression of alloantibody.
for the treatment of early relapsing–remitting multiple Rituximab (6), a chimeric (mouse and human) CD20-specific mAb, markedly
sclerosis was associated with the development of auto‑ depletes levels of B lymphocytes. Abbreviations: BAFF-R, BAFF receptor; BCR, B-cell
antibodies, predominantly against thyroid antigens and receptor; BCMA, B-cell maturation antigen; IVIG, intravenous immunoglobulin; mAb,
blood products, in 30% of patients.108–110 In summary, monoclonal antibody; MHC, major histocompatibility complex; TCR, T-cell receptor;
the simple concept that partial B‑cell depletion mediated TNF, tumor necrosis factor; TNFRSF13B, TNF receptor superfamily member 13B;
TNFRSF13C, TNF receptor superfamily member 13C; TNFSF13, TNF ligand
by alemtuzumab can reduce B‑cell function might be
superfamily member 13; TNFSF13B, TNF ligand superfamily member 13B.
fundamentally flawed.
T h e p o l y c l o n a l r a b b i t AT G p r e p a r a t i o n ,
Thymoglobulin® , contains antibody molecules with and B cells has been associated with increased produc‑
specificity for many cell surface proteins including tion of TnFsF13B, perhaps in response to homeostatic
those found on B cells and T cells. Thymoglobulin® as pressure to replace the depleted lymphocytes. Increased
an induction therapy in renal transplant recipients effec‑ TnFsF13B production might lead to paradoxical B‑cell
tively reverses early renal transplant rejection. The effect activation in the setting of B‑cell lymphopenia.114
of Thymoglobulin® on early and late AMR is unclear. since the TnFsF13B‑mediated processes that regulate
B‑cell survival are critical for efficient B‑cell‑mediated
TNfSf13B inhibition immune responses, members of the TnF ligand super‑
TnFsF13B is a member of the TnF superfamily of pro‑ family have emerged as potential key players in the
teins. It is a secreted cytokine that, along with TnFsF13 etiology and treatment of autoimmunity and AMR.
(formally termed APRIl), interacts with three recep‑ Modulating B‑cell homeostasis and survival via targeting
tors expressed on the surface of B cells: TnFRsF13B, of TnFsF13B has potentially important implications for
TnFRsF17 and, most importantly, TnFRsF13C (form‑ transplantation tolerance. For example, ablation of B‑cell
erly termed TACI, BCMA and the BAFF receptor, respec‑ clones in combination with controlled repopulation
tively). Integrated signals from both the B‑cell receptors using TnF ligand superfamily inhibitors might facilitate
and members of the TnF ligand superfamily are essen‑ the induction of transplantation tolerance, because the
tial for the establishment and maintenance of B‑cell newly emerged B cells could undergo induction of tolero‑
clones.111,112 Together with B‑cell receptor signaling, TnF‑ genic mechanisms in response to exposure to donor
ligand‑superfamily signaling transduces survival signals antigens (Figure 2).
that determine the proportion of newly formed B cells Currently, at least three agents that inhibit the inter‑
that survive to maturity, the longevity of mature primary action of TnFsF13B with receptors on the surface of
B cells, and the differentiation of antigen‑exposed subsets B cells are being developed. Belimumab is a fully human
of B cells.26,46,113 Moreover, substantial depletion of T cells monoclonal antibody against TnFsF13B. Atacicept is a

nATuRe ReVIews | NEphRology VoluMe 6 | oCToBeR 2010 | 589


© 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

fusion protein that combines the extracellular ligand‑ positivity for donor‑specific antibodies than patients
binding portion of TnFRsF13B with an Fc region of IgG treated with ciclosporin.124 efforts to inhibit the pathway
to inhibit binding to TnFsF13B and TnFsF13. BR3‑Fc associated with the CD40–CD154 interaction have been
is a recombinant fusion protein that utilizes the ligand‑ uniformly successful in achieving transplantation toler‑
binding portion of TnFRsF13C to prevent its binding to ance in animal models. Interestingly, despite the clear
TnFsF13B (Figure 2). role of CD40 stimulation in B‑cell activation and in B‑cell
class switching, blockade of the CD40–CD154 interac‑
Co-stimulation blockade tion does not seem to inhibit alloantibody production.125
The ‘help’ provided by T cells to antigen‑specific naive The reasons for this observation are unclear (Figure 2).
B cells in the form of co‑stimulatory signals and cyto‑ several other B‑cell co‑stimulatory interactions have
kines is essential to the formation of antibody‑secreting been defined, such as the CTlA‑4–ICos ligand inter‑
cells.115,116 Transplant rejection processes mediated by action, the CD27–CD70 interaction and the CD134–
T cells and by B cells are not, therefore, mutually exclu‑ oX40l interaction.126 The effects of blockade of these
sive. However, in anamnestic immune responses (which interactions should be tested in rigorous animal models
involve the prompt reappearance of antibodies after of allograft transplantation.
rechallenge with an antigen that had previously induced a
primary immune response) the extent of interdependency Spontaneous renal transplant tolerance
of T‑cell and B‑cell responses decreases. Reports of cases of spontaneous allograft tolerance
Co‑stimulatory molecules are cell‑surface receptors demon strate that transplantation tolerance can be
that provide necessary accessory signals at the time achieved. Generally, these spontaneous phenomena
of antigen–receptor interaction to shape the magni‑ have been detected in patients who were nonadherent to
tude and character of the subsequent antigen‑specific immunosuppressive therapy or who required immuno‑
response. Blockade of T‑cell co‑stimulatory molecules suppression withdrawal owing to complications, both of
has been effectively targeted to achieve T‑cell tolerance which commonly lead to organ rejection. The proportion
to allograft transplantation, and this approach also pre‑ of kidney transplant recipients reported as spontane‑
vents the formation of alloantigen‑specific antibodies ously tolerant is small (<1% of all renal transplant recipi‑
and auto antibodies. 117–120 Most co‑stimulatory mol‑ ents), so performing a systematic mechanistic analysis
ecules have been identified on the basis of their influ‑ in these patients has not been possible. However, a reg‑
ence on T cells; B‑cell‑specific co‑stimulatory molecules istry established by the Immune Tolerance network has
have also been identified, but as yet they have not been been used to conduct an exploratory mechanistic study.
used therapeutically. The investigators identified 25 kidney transplant recipi‑
The receptor–co‑stimulator interaction pathways ents who were considered transplant tolerant, defined
CD28–CD80, CD28–CD86 and CD154–CD40 have as having stable graft function and having received no
been extensively studied in animal models of transplanta‑ immunosuppression for at least 1 year. An analysis of the
tion. The extent to which these pathways influence B‑cell gene‑expression profiles in subsets of peripheral blood
function through control of T‑cell help is hard to define. lymphocytes from these patients, from patients with
Given that anamnestic responses become decreasingly stable graft function who were on immunosuppression,
susceptible to co‑stimulatory blockade, it is likely that and from healthy controls, suggested that allograft toler‑
much of their influence is T‑cell related. nevertheless, ance was strongly associated with an increased expres‑
these pathways clearly influence antibody formation and sion of multiple genes involved in B‑cell differentiation.
are involved in the presentation of antigens by B cells to This subset of B‑cell‑related genes or ‘B‑cell signature’
T cells. Co‑stimulation blockade (that is, blockade of was consistent with the upregulation of CD20 mes‑
CD28–B7.1, CD28–B7.2 and/or CD154–CD40 interaction senger RnA in urine sediment cells and the elevated
pathways) has been used in several murine and non‑ numbers of naive and transitional B cells as assessed by
human primate models of allograft rejection to induce flow cytometric analysis in spontaneously transplant‑
tolerogenic mechanisms that are active during peripheral tolerant kidney transplant recipients. 127 similarly, a
antigen encounters in graft rejection, either in animals study from the Indices of Tolerance european union
with an allograft alone or in animals with an allograft and consortium, which analogously screened 11 opera‑
transfused hematopoietic cells (Figure 2).66,69,121 tionally tolerant kidney transplant recipients, cohorts
Abatacept, a fusion protein that competes with CD28 of immunosuppressed recipients exhibiting allograft
for binding with CD80 (B7.1) and CD86 (B7.2), impedes injury and healthy controls, found that peripheral
the formation of autoantibodies. Belatacept, a derivative blood from tolerant patients contained higher levels
of abatacept with increased affinity for CD80 and CD86, of B‑cell‑related genes than peripheral blood from the
was developed specifically for use in the organ trans‑ other patients.128 These results suggest a critical role for
plantation setting, where robust immunosuppression B cells in the regulation of the alloimmune response in
is required.122,123 Both belatacept and abatacept inhibit transplant‑tolerant kidney graft recipients. In addition,
de novo formation of antibodies (including allo‑ the results provide a candidate set of genes that may
antibodies) in non‑primates.119 The results of a random‑ serve as biomarkers to identify renal transplant recipi‑
ized clinical trial in renal transplant recipients showed ents who may benefit from minimization or withdrawal
that patients treated with belatacept have lower rates of of immunosuppression.

590 | OCTOBER 2010 | vOlumE 6 www.nature.com/nrneph


© 2010 Macmillan Publishers Limited. All rights reserved
f o C u S o N To l E R A N C E I N T R A N S p l A N TAT I o N

Conclusions will be able to integrate them safely into existing


T‑cell‑mediated allograft rejection is generally well immunosuppressive regimens.
controlled and understood. However, alloantigen‑
specific B‑cell biology is less well understood than Review criteria
T‑cell biology, and B‑cell‑mediated humoral immune We searched the MEDLINE database for papers in
responses are only now being recognized as a significant English published between 1970 and 2009 (inclusive),
barrier to developing allograft tolerance. Many thera‑ using the terms: “B-cell tolerance”, “antibody-mediated
pies are emerging that have the ability to control B‑cell‑ rejection”, “neonatal tolerance”, “mixed chimerism”,
mediated immune responses. ultimately, the clinical “alemtuzumab”, “rituximab”, “eculizumab”, “bortezomib”,
efficacy of these strategies will depend on their ability “BAFF and transplantation”, “co-stimulation blockade and
to shape the function and identity of B‑cell subsets, transplantation”. The reference lists of retrieved articles
were also searched to identify relevant papers.
and whether the research and clinical communities

1. Meier-Kriesche, H. U., Schold, J. D. & Kaplan, B. 17. Wardemann, H. et al. Predominant autoantibody 33. Cooke, M. P. et al. Immunoglobulin signal
Long-term renal allograft survival: have we made production by early human B cell precursors. transduction guides the specificity of B cell-T cell
significant progress or is it time to rethink our Science 301, 1374–1377 (2003). interactions and is blocked in tolerant self-
analytic and therapeutic strategies? Am. J. 18. Burnet, F. M. & Fenner, F. The Production of reactive B cells. J. Exp. Med. 179, 425–438
Transplant. 4, 1289–1295 (2004). Antibodies 2nd edn (Macmillan, Melbourne, (1994).
2. Meier-Kriesche, H. U., Schold, J. D., Srinivas, T. R. 1949). 34. Erikson, J. et al. Expression of anti-DNA
& Kaplan, B. Lack of improvement in renal 19. Goodnow, C. C. et al. Altered immunoglobulin immunoglobulin transgenes in non-autoimmune
allograft survival despite a marked decrease in expression and functional silencing of self- mice. Nature 349, 331–334 (1991).
acute rejection rates over the most recent era. reactive B lymphocytes in transgenic mice. 35. Merrell, K. T. et al. Identification of anergic
Am. J. Transplant. 4, 378–383 (2004). Nature 334, 676–682 (1988). B cells within a wild-type repertoire. Immunity
3. Venetz, J. P. & Pascual, M. New treatments for 20. Nemazee, D. A. & Bürki, K. Clonal deletion of 25, 953–962 (2006).
acute humoral rejection of kidney allografts. B lymphocytes in a transgenic mouse bearing 36. Russell, D. M. et al. Peripheral deletion of self-
Expert Opin. Investig. Drugs 16, 625–633 (2007). anti-MHC class I antibody genes. Nature 337, reactive B cells. Nature 354, 308–311 (1991).
4. Moll, S. & Pascual, M. Humoral rejection of organ 562–566 (1989). 37. Bell, S. E. & Goodnow, C. C. A selective defect in
allografts. Am. J. Transplant. 5, 2611–2618 21. Hartley, S. B. et al. Elimination of self-reactive IgM antigen receptor synthesis and transport
(2005). B lymphocytes proceeds in two stages: causes loss of cell surface IgM expression on
5. Feucht, H. E. Significance of donor-specific arrested development and cell death. Cell 72, tolerant B lymphocytes. EMBO J. 13, 816–826
antibodies in acute rejection. Transplant. Proc. 325–335 (1993). (1994).
37, 3693–3694 (2005). 22. Allman, D. et al. Resolution of three 38. Rui, L., Healy, J. I., Blasioli, J. & Goodnow, C. C.
6. Feucht, H. E. & Mihatsch, M. J. Diagnostic value nonproliferative immature splenic B cell subsets ERK signaling is a molecular switch integrating
of C4d in renal biopsies. Curr. Opin. Nephrol. reveals multiple selection points during opposing inputs from B cell receptor and T cell
Hypertens. 14, 592–598 (2005). peripheral B cell maturation. J. Immunol. 167, cytokines to control TLR4-driven plasma cell
7. Racusen, L. C. et al. Antibody-mediated rejection 6834–6840 (2001). differentiation. J. Immunol. 177, 5337–5346
criteria—an addition to the Banff 97 23. Lang, J. et al. Enforced Bcl-2 expression inhibits (2006).
classification of renal allograft rejection. Am. J. antigen-mediated clonal elimination of peripheral 39. Lesley, R. et al. Reduced competitiveness of
Transplant. 3, 708–714 (2003). B cells in an antigen dose-dependent manner autoantigen-engaged B cells due to increased
8. Colvin, R. B. & Smith, R. N. Antibody-mediated and promotes receptor editing in autoreactive, dependence on BAFF. Immunity 20, 441–453
organ-allograft rejection. Nat. Rev. Immunol. 5, immature B cells. J. Exp. Med. 186, 1513–1522 (2004).
807–817 (2005). (1997). 40. Aplin, B. D. et al. Tolerance through indifference:
9. Stegall, M. D., Dean, P. G. & Gloor, J. Mechanisms 24. Fang, W. et al. CD40 inhibits B cell apoptosis by autoreactive B cells to the nuclear antigen La
of alloantibody production in sensitized renal upregulating bcl-xL expression and blocking show no evidence of tolerance in a transgenic
allograft recipients. Am. J. Transplant. 9, oxidant accumulation. Am. J. Physiol. 272, model. J. Immunol. 171, 5890–5900 (2003).
998–1005 (2009). C950–C956 (1997). 41. Hannum, L. G., Ni, D., Haberman, A. M.,
10. Cornell, L. D., Smith, R. N. & Colvin, R. B. Kidney 25. Strasser, A. & Bouillet, P. The control of Weigert, M. G. & Shlomchik, M. J. A disease-
transplantation: mechanisms of rejection and apoptosis in lymphocyte selection. Immunol. related rheumatoid factor autoantibody is not
acceptance. Annu. Rev. Pathol. 3, 189–220 Rev. 193, 82–92 (2003). tolerized in a normal mouse: implications for the
(2008). 26. Mackay, F. & Schneider, P. Cracking the BAFF origins of autoantibodies in autoimmune
11. Vongwiwatana, A., Tasanarong, A., Hidalgo, L. G. code. Nat. Rev. Immunol. 9, 491–502 (2009). disease. J. Exp. Med. 184, 1269–1278 (1996).
& Halloran, P. F. The role of B cells and 27. Tiegs, S. L., Russell, D. M. & Nemazee, D. 42. Nemazee, D. Mechanisms and meaning of
alloantibody in the host response to human Receptor editing in self-reactive bone B-lymphocyte tolerance. Res. Immunol. 143,
organ allografts. Immunol. Rev. 196, 197–218 marrow B cells. J. Exp. Med. 177, 1009–1020 272–275 (1992).
(2003). (1993). 43. Cyster, J. G., Hartley, S. B. & Goodnow, C. C.
12. Goodnow, C. C., Sprent, J., Fazekas de St 28. Gay, D., Saunders, T., Camper, S. & Weigert, M. Competition for follicular niches excludes self-
Groth, B. & Vinuesa, C. G. Cellular and genetic Receptor editing: an approach by autoreactive reactive cells from the recirculating B-cell
mechanisms of self tolerance and autoimmunity. B cells to escape tolerance. J. Exp. Med. 177, repertoire. Nature 371, 389–395 (1994).
Nature 435, 590–597 (2005). 999–1008 (1993). 44. Stadanlick, J. E. & Cancro, M. P. BAFF and the
13. Shlomchik, M. J. Sites and stages of autoreactive 29. Casellas, R. et al. Contribution of receptor plasticity of peripheral B cell tolerance. Curr.
B cell activation and regulation. Immunity 28, editing to the antibody repertoire. Science 291, Opin. Immunol. 20, 158–161 (2008).
18–28 (2008). 1541–1544 (2001). 45. Stadanlick, J. E. et al. Tonic B cell antigen
14. Nemazee, D. Does immunological tolerance 30. Hippen, K. L. et al. In vivo assessment of the receptor signals supply an NF-kappaB substrate
explain the waste in the B-lymphocyte immune relative contributions of deletion, anergy, and for prosurvival BLyS signaling. Nat. Immunol. 9,
system? Experiment and theory. Ann. NY Acad. editing to B cell self-tolerance. J. Immunol. 175, 1379–1387 (2008).
Sci. 764, 397–401 (1995). 909–916 (2005). 46. Treml, L. S., Quinn, W. J. 3rd, Treml, J. F.,
15. Nemazee, D. Receptor editing in lymphocyte 31. Goodnow, C. C. Cellular mechanisms of self- Scholz, J. L. & Cancro, M. P. Manipulating B cell
development and central tolerance. Nat. Rev. tolerance. Curr. Opin. Immunol. 2, 226–236 homeostasis: a key component in the
Immunol. 6, 728–740 (2006). (1989). advancement of targeted strategies. Arch.
16. Pelanda, R. et al. Receptor editing in a transgenic 32. Goodnow, C. C., Crosbie, J., Jorgensen, H., Immunol. Ther. Exp. (Warsz) 56, 153–164
mouse model: site, efficiency, and role in B cell Brink, R. A. & Basten, A. Induction of self- (2008).
tolerance and antibody diversification. Immunity tolerance in mature peripheral B lymphocytes. 47. Glotz, D., Antoine, C. & Duboust, A. Antidonor
7, 765–775 (1997). Nature 342, 385–391 (1989). antibodies and transplantation: how to deal with

nATuRe ReVIews | NEphRology VoluMe 6 | oCToBeR 2010 | 591


© 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS
them before and after transplantation. unmet needs. Am. J. Transplant. 6, 884–893 and renal transplantation for multiple myeloma
Transplantation 79 (Suppl.), S30–S32 (2005). (2006). with end stage renal disease: the induction of
48. Cerny, T., Borisch, B., Introna, M., Johnson, P. & 67. Kirk, A. D. Crossing the bridge: large animal allograft tolerance through mixed
Rose, A. L. Mechanism of action of rituximab. models in translational transplantation lymphohematopoietic chimerism.
Anticancer Drugs 13 (Suppl. 2), S3–S10 (2002). research. Immunol. Rev. 196, 176–196 (2003). Transplantation 68, 480–484 (1999).
49. Aranda, J. M. Jr et al. Anti-CD20 monoclonal 68. Lechler, R. I., Sykes, M., Thomson, A. W. & 86. Fudaba, Y. et al. Myeloma responses and
antibody (rituximab) therapy for acute cardiac Turka, L. A. Organ transplantation—how much of tolerance following combined kidney and
humoral rejection: a case report. Transplantation the promise has been realized? Nat. Med. 11, monmyeloablative marrow transplantation:
73, 907–910 (2002). 605–613 (2005). in vivo and in vitro analyses. Am. J. Transplant.
50. Garrett, H. E. Jr, Groshart, K., Duvall-Seaman, D., 69. Sykes, M. Immune tolerance: mechanisms and 6, 2121–2133 (2006).
Combs, D. & Suggs, R. Treatment of humoral application in clinical transplantation. J. Intern. 87. Kawai, T. et al. HLA-mismatched renal
rejection with rituximab. Ann. Thorac. Surg. 74, Med. 262, 288–310 (2007). transplantation without maintenance
1240–1242 (2002). 70. Owen, R. D. Immunogenetic consequences of immunosuppression. N. Engl. J. Med. 358,
51. Becker, Y. T., Becker, B. N., Pirsch, J. D. & vascular anastomoses between bovine twins. 353–361 (2008).
Sollinger, H. W. Rituximab as treatment for Science 102, 400–401 (1945). 88. Porcheray, F. et al. B-cell immunity in the context
refractory kidney transplant rejection. Am. J. 71. Billingham, R. E., Brent, L. & Medawar, P. B. of T-cell tolerance after combined kidney and
Transplant. 4, 996–1001 (2004). Actively acquired tolerance of foreign cells. bone marrow transplantation in humans. Am. J.
52. Sonnenday, C. J. et al. Plasmapheresis, CMV Nature 172, 603–606 (1953). Transplant. 9, 2126–2135 (2009).
hyperimmune globulin, and anti-CD20 allow ABO- 72. Rieben, R., Tucci, A., Nydegger, U. E. & 89. Page, A., Kirk, A. D., Larsen, C. & Kean L. S.
incompatible renal transplantation without Zubler, R. H. Self tolerance to human A and B Prolonged engraftment but breakthrough
splenectomy. Am. J. Transplant. 4, 1315–1322 histo-blood group antigens exists at the B cell rejection during primate mixed chimerism
(2004). level and cannot be broken by potent polyclonal induction: T cell alloreactivity remains a barrier
53. Vieira, C. A. et al. Rituximab for reduction of anti- B cell activation in vitro. Eur. J. Immunol. 22, to tolerance despite full MHC matching. Am. J.
HLA antibodies in patients awaiting renal 2713–2717 (1992). Transplant. 10, 146 (2010).
transplantation: 1. Safety, pharmacodynamics, 73. Fong, S. W., Qaqundah, B. Y. & Taylor, W. F. 90. Terasaki, P. I. Humoral theory of
and pharmacokinetics. Transplantation 77, Developmental patterns of ABO isoagglutinins in transplantation. Am. J. Transplant. 3, 665–673
542–548 (2004). normal children correlated with the effects of (2003).
54. Gloor, J. M. et al. Overcoming a positive age, sex, and maternal isoagglutinins. 91. Calne, R. Y. Prope tolerance: the future of organ
crossmatch in living-donor kidney Transfusion 14, 551–559 (1974). transplantation--from the laboratory to the
transplantation. Am. J. Transplant. 3, 74. West, L. J. et al. ABO-incompatible heart clinic. Transplantation 77, 930–932 (2004).
1017–1023 (2003). transplantation in infants. N. Engl. J. Med. 344, 92. Kirk, A. D. et al. Results from a human renal
55. Tydén, G. et al. ABO incompatible kidney 793–800 (2001). allograft tolerance trial evaluating the
transplantations without splenectomy, using 75. West, L. J. et al. Outcomes of the world humanized CD52-specific monoclonal antibody
antigen-specific immunoadsorption and experience in ABO-compatible infant heart alemtuzumab (CAMPATH-1H). Transplantation
rituximab. Am. J. Transplant. 5, 145–148 (2005). transplantation. Am. J. Transplant. 5, 157 76, 120–129 (2003).
56. Richardson, P. G. et al. Bortezomib or high-dose (2005). 93. Ciancio, G. et al. The use of Campath-1H as
dexamethasone for relapsed multiple myeloma. 76. West, L. J. Targeting antibody-mediated rejection induction therapy in renal transplantation:
N. Engl. J. Med. 352, 2487–2498 (2005). in the setting of ABO-incompatible infant heart preliminary results. Transplantation 78,
57. Vanderlugt, C. L., Rahbe, S. M., Elliott, P. J., transplantation: graft accommodation vs. B cell 426–433 (2004).
Dal Canto, M. C. & Miller, S. D. Treatment of tolerance. Curr. Drug Targets Cardiovasc. 94. Calne, R. et al. Prope tolerance, perioperative
established relapsing experimental autoimmune Haematol. Disord. 5, 223–232 (2005). campath 1H, and low-dose cyclosporin
encephalomyelitis with the proteasome inhibitor 77. Fan, X. et al. Donor-specific B-cell tolerance after monotherapy in renal allograft recipients.
PS-519. J. Autoimmun. 14, 205–211 (2000). ABO-incompatible infant heart transplantation. Lancet 351, 1701–1702 (1998).
68. Neubert, K. et al. The proteasome inhibitor Nat. Med. 10, 1227–1233 (2004). 95. Calne, R. et al. Campath IH allows low-dose
bortezomib depletes plasma cells and protects 78. Gugenheim, J., Samuel, D., Reynes, M. & cyclosporine monotherapy in 31 cadaveric renal
mice with lupus-like disease from nephritis. Nat. Bismuth, H. Liver transplantation across ABO allograft recipients. Transplantation 68,
Med. 14, 748–755 (2008). blood group barriers. Lancet 336, 519–523 1613–1616 (1999).
59. Ron, D. & Walter, P. Signal integration in the (1990). 96. Ciancio, G. & Burke, G. W. 3rd. Alemtuzumab
endoplasmic reticulum unfolded protein 79. Stegall, M. D., Dean, P. G. & Gloor, J. M. ABO- (Campath-1H) in kidney transplantation. Am. J.
response. Nat. Rev. Mol. Cell Biol. 8, 519–529 incompatible kidney transplantation. Transplant. 8, 15–20 (2008).
(2007). Transplantation 78, 635–640 (2004). 97. Kirk, A. D. et al. Results from a human renal
60. Perry, D. K. et al. Proteasome inhibition causes 80. Ildstad, S. T. & Sachs, D. H. Reconstitution with allograft tolerance trial evaluating T-cell
apoptosis of normal human plasma cells syngeneic plus allogeneic or xenogeneic bone depletion with alemtuzumab combined with
preventing alloantibody production. Am. J. marrow leads to specific acceptance of deoxyspergualin. Transplantation 80,
Transplant. 9, 201–209 (2009). allografts or xenografts. Nature 307, 168–170 1051–1059 (2005).
61. Everly, M. J. et al. Bortezomib provides effective (1984). 98. Knechtle, S. J. et al. Campath-1H in renal
therapy for antibody- and cell-mediated acute 81. Sharabi, Y. & Sachs, D. H. Mixed chimerism and transplantation: The University of Wisconsin
rejection. Transplantation 86, 1754–1761 permanent specific transplantation tolerance experience. Surgery 136, 754–760 (2004).
(2008). induced by a nonlethal preparative regimen. 99. Vathsala, A. et al. Randomized trial of
62. Trivedi, H. L. et al. Abrogation of anti-HLA J. Exp. Med. 169, 493–502 (1989). alemtuzumab for prevention of graft rejection
antibodies via proteasome inhibition. 82. Ildstad, S. T., Wren, S. M., Bluestone, J. A., and preservation of renal function after kidney
Transplantation 87, 1555–1561 (2009). Barbieri, S. A. & Sachs, D. H. Characterization transplantation. Transplantation 80, 765–774
63. Sberro-Soussan, R. et al. Bortezomib as the sole of mixed allogeneic chimeras. (2005).
post-renal transplantation desensitization agent Immunocompetence, in vitro reactivity, and 100. Watson, C. J. et al. Alemtuzumab (CAMPATH 1H)
does not decrease donor-specific anti-HLA genetic specificity of tolerance. J. Exp. Med. 162, induction therapy in cadaveric kidney
antibodies. Am. J. Transplant. 10, 681–686 231–244 (1985). transplantation—efficacy and safety at five
(2010). 83. Domenig, C. et al. Roles of deletion and years. Am. J. Transplant. 5, 1347–1353 (2005).
64. Locke, J. E. et al. The use of antibody to regulation in creating mixed chimerism and 101. Ciancio, G. et al. A randomized trial of three
complement protein C5 for salvage treatment of allograft tolerance using a nonlymphoablative renal transplant induction antibodies:
severe antibody-mediated rejection. Am. J. irradiation-free protocol. J. Immunol. 175, 51–60 early comparison of tacrolimus, mycophenolate
Transplant. 9, 231–235 (2009). (2005). mofetil, and steroid dosing, and newer immune-
65. Stegall, M. D., Diwan, T. S. & Burns, J. M. 84. Millan, M. T. et al. Mixed chimerism and monitoring. Transplantation 80, 457–465
Prevention of acute humoral rejection with C5 immunosuppressive drug withdrawal after HLA- (2005).
inhibition. Am. J. Transplant. 9 (Suppl. 2), mismatched kidney and hematopoietic 102. Flechner, S. M. et al. Alemtuzumab induction
241–242 (2009). progenitor transplantation. Transplantation 73, and sirolimus plus mycophenolate mofetil
66. Kean, L. S., Gangappa, S., Pearson, T. C. & 1386–1391 (2002). maintenance for CNI and steroid-free kidney
Larsen, C. P. Transplant tolerance in non-human 85. Spitzer, T. R. et al. Combined histocompatibility transplant immunosuppression. Am. J.
primates: progress, current challenges and leukocyte antigen-matched donor bone marrow Transplant. 5, 3009–3014 (2005).

592 | OCTOBER 2010 | vOlumE 6 www.nature.com/nrneph


© 2010 Macmillan Publishers Limited. All rights reserved
f o C u S o N To l E R A N C E I N T R A N S p l A N TAT I o N
103. Tan, H. P. et al. Steroid-free tacrolimus 112. Treml, J. F., Hao, Y., Stadanlick, J. E. & 121. Larsen, C. P., Knechtle, S. J., Adams, A.,
monotherapy after pretransplantation Cancro, M. P. The BLyS family: toward a Pearson, T. & Kirk, A. D. A new look at blockade of
thymoglobulin or Campath and laparoscopy in molecular understanding of B cell homeostasis. T-cell costimulation: a therapeutic strategy for
living donor renal transplantation. Transplant. Cell Biochem. Biophys. 53, 1–16 (2009). long-term maintenance immunosuppression. Am.
Proc. 37, 4235–4240 (2005). 113. Cancro, M. P. Signalling crosstalk in B cells: J. Transplant. 6, 876–883 (2006).
104. Kaufman, D. B. et al. Alemtuzumab induction managing worth and need. Nat. Rev. Immunol. 9, 122. Vincenti, F. Costimulation blockade in
and prednisone-free maintenance 657–661 (2009). autoimmunity and transplantation. J. Allergy Clin.
immunotherapy in kidney transplantation: 114. Bloom, D. et al. BAFF is increased in renal Immunol. 121, 299–306 (2008).
comparison with basiliximab induction— transplant patients following treatment with 123. Vincenti, F. & Kirk, A. D. What’s next in the
long-term results. Am. J. Transplant. 5, alemtuzumab. Am. J. Transplant. 9, 1835–1845 pipeline. Am. J. Transplant. 8, 1972–1981 (2008).
2539–2548 (2005). (2009). 124. Vincenti, F. et al. A phase III study of belatacept-
105. Margreiter, R. et al. Alemtuzumab (Campath-1H) 115. Claman, H. N., Chaperon, E. A. & Triplett, R. F. based immunosuppression regimens versus
and tacrolimus monotherapy after renal Thymus-marrow cell combinations. Synergism in cyclosporine in renal transplant recipients
transplantation: results of a prospective antibody production. Proc. Soc. Exp. Biol. Med. (BENEFIT study). Am. J. Transplant. 10, 535–546
randomized trial. Am. J. Transplant. 8, 122, 1167–1171 (1966). (2010).
1480–1485 (2008). 116. Miller, J. F. & Mitchell, G. F. Cell to cell interaction 125. Kirk, A. D. et al. Treatment with humanized
106. Vo, A. A. et al. Analysis of subcutaneous (SQ) in the immune response. I. Hemolysin-forming monoclonal antibody against CD154 prevents
alemtuzumab induction therapy in highly cells in neonatally thymectomized mice acute renal allograft rejection in nonhuman
sensitized patients desensitized with IVIG and reconstituted with thymus or thoracic duct primates. Nat. Med. 5, 686–693 (1999).
rituximab. Am. J. Transplant. 8, 144–149 (2008). lymphocytes. J. Exp. Med. 128, 801–820 126. Li, X. C., Rothstein, D. M. & Sayegh, M. H.
107. Pascual, J. et al. Alemtuzumab induction and (1968). Costimulatory pathways in transplantation:
antibody-mediated kidney rejection after 117. Davidson, A. et al. Co-stimulatory blockade in the challenges and new developments. Immunol.
simultaneous pancreas-kidney transplantation. treatment of murine systemic lupus Rev. 229, 271–293 (2009).
Transplantation 87, 125–132 (2009). erythematosus (SLE). Ann. NY Acad. Sci. 987, 127. Newell, K. A. et al. Identification of a B cell
108. Jones, J. L. & Coles, A. J. Spotlight on 188–198 (2003). signature associated with renal transplant
alemtuzumab. Int. MS J. 16, 77–81 (2009). 118. Finck, B. K., Linsley, P. S. & Wofsy, D. Treatment tolerance in humans. J. Clin. Invest. 120,
109. Jones, J. L. et al. IL-21 drives secondary of murine lupus with CTLA4Ig. Science 265, 1836–1847 (2010).
autoimmunity in patients with multiple sclerosis, 1225–1227 (1994). 128. Sagoo, P. et al. Development of a cross-platform
following therapeutic lymphocyte depletion with 119. Cardona, K. et al. Long-term survival of neonatal biomarker signature to detect renal transplant
alemtuzumab (Campath-1H). J. Clin. Invest. 119, porcine islets in nonhuman primates by targeting tolerance in humans. J. Clin. Invest. 120,
2052–2061 (2009). costimulation pathways. Nat. Med. 12, 304–306 1848–1861 (2010).
110. Coles, A. J. et al. Alemtuzumab vs. interferon (2006).
beta-1a in early multiple sclerosis. N. Engl. J. 120. Ford, M. L. et al. A critical precursor frequency of Author contributions
Med. 359, 1786–1801 (2008). donor-reactive CD4+ T cell help is required for A. D. Kirk, N. A. Turgeon and N. N. Iwakoshi
111. Crowley, J. E. et al. Homeostatic control of CD8+ T cell-mediated CD28/CD154- contributed equally to researching data, discussion of
B lymphocyte subsets. Immunol. Res. 42, independent rejection. J. Immunol. 180, content, writing, and reviewing/editing the manuscript
75–83 (2008). 7203–7211 (2008). before submission.

nATuRe ReVIews | NEphRology VoluMe 6 | oCToBeR 2010 | 593


© 2010 Macmillan Publishers Limited. All rights reserved

Potrebbero piacerti anche