Monitoring Stem Cell Research
The President's Council on Bioethics
Washington, D.C.
January 2004 www.bioethics.gov
Pre-Publication Version
Appendix L Stem Cells and Tissue Regeneration:
Lessons From Recipients of Solid Organ Transplantation
Silviu Itescu M.D.
Director of Transplantation Immunology,
Departments of Medicine and Surgery,
Columbia University, New York, NY
CONTENTS
1. OVERVIEW
2. IMMUNOBIOLOGY OF ORGANTRANSPLANTATION
The Human Leukocyte Antigens (HLA)
T Cell Recognition Of Antigen Presented By HLA Molecules
Thymic Education Of T Cells
T Cell Recognition Of Alloantigens
Tolerance Induction
3. IMMUNOSUPPRESSIVE AGENTS COMMONLY USED IN ORGAN TRANSPLANT
RECIPIENTS: BENEFITS AND ADVERSE OUTCOMES
4. STEM CELL TRANSPLANTATION AND IMMUNOSUPPRESSION
Materno-Fetal Tolerance
Immunogenic Characteristics Of Embryonic And Adult Stem
Cells
Tolerogenic Effects Of Stem Cell Transplantation
1. OVERVIEW
The Major Histocompatibility Complex (MHC) is located on
the short arm of chromosome 6 in humans and encodes the alloantigens
known as Human Leukocyte Antigens (HLA), polymorphic cell
surface molecules which enable the immune system to recognize
both self and foreign antigens. The class II HLA molecules
(HLA-DR, HLA-DP, and HLA-DQ) are usually found only on antigen-presenting
cells such as B lymphocytes, macrophages, and dendritic cells
of lymphoid organs, and initiate the immune response to foreign
proteins, including viruses, bacteria, and foreign HLA antigens
on transplanted organs.
Following binding of foreign proteins, class II HLA on antigen-presenting
cells activate CD4+ T cells, which in turn activate cytotoxic
CD8+ T cells to recognize the same foreign antigen bound to
HLA class I (HLA-A, HLA-B, and HLA-C, molecules found on the
surface of all cells) and destroy the target. The actual recognition
of foreign HLA transplantation antigens by T cells is referred
to as allorecognition. Two distinct pathways of allorecognition
have been described, direct and indirect. The direct pathway
involves receptors on the host T cells that directly recognize
intact HLA antigens on the cells of the transplanted organ.
The indirect pathway requires an antigen-presenting cell that
internalizes the foreign antigen and presents it via its own
HLA class II molecule on the surface of an antigen-presenting
cell to the CD4+ helper T cells.
Once recognition has taken place, an important cascade of
events is initiated at the cellular level, culminating in
intracellular release of ionized calcium from intracellular
stores. The calcium binds with a regulatory protein called
calmodulin, forming a complex that activates various phosphatases,
particularly calcineurin. Calcineurin dephosphorylates an
important cytoplasmic protein called nuclear factor of activated
T cells (NFAT), resulting in its migration to the nucleus
and induction of the production of various cytokines such
as IL-2. These cytokines recruit other T cells to destroy
the transplanted organ, ultimately resulting in rejection
and loss of the graft.
Immunosuppressive regimens used to prevent allograft rejection
are aimed at inhibiting the various arms of the immune response,
typically require multiagent combinations, and need to be
maintained for the duration of life. The currently used armamentarium
confers significant side-effect risks, including infectious
and neoplastic complications. Moreover, despite success at
preventing early allograft rejection, long-term survival of
transplanted organs remains difficult to achieve and novel
methods to achieve long-term tolerance are being actively
sought.
Stem cells obtained from embryonic or adult sources differ
from other somatic cells in that they express very low levels
of HLA molecules on their cell surfaces. This endows these
cell types with the theoretical potential to escape the standard
mechanisms of immune rejection discussed above. However, under
conditions that enable cellular differentiation in vitro and
in vivo each of these stem cell populations acquires high
level expression of HLA molecules, suggesting that their long-term
survival following transplantation in vivo may be limited
by typical immune rejection phenomena. Recent experimental
data, however, provide striking counterintuitive examples
that stem cells from both embryonic and adult sources may
evade the recipient's immune system and result in long-term
engraftment in the absence of immunosuppression despite acquisition
of surface HLA molecule expression. These observations may
have significant impact on the emerging field of regenerative
medicine.
2. IMMUNOBIOLOGY OF ORGAN TRANSPLANTATION
The Human Leukocyte Antigens (HLA)
Differences between individuals which enable immune recognition
of non-self from self are principally due to the extreme polymorphism
of genes in the Major Histocompatibility Complex (MHC) on
chromosome 6 in man which encode the cell surface HLA molecules.
These molecules are cell surface glycoproteins whose biologic
function is to bind antigenic peptides (epitopes) derived
from viruses, bacteria, or cancer cells, and present them
to T cells for subsequent immune recognition. Each HLA gene
includes a large number of alleles and the peptide binding
specificity varies for each different HLA allele. The 1996
WHO HLA Nomenclature Committee report lists more than 500
different HLA class I and class II alleles.
Crystallographic x-ray studies have demonstrated that the
hypervariable regions encoded by polymorphic regions in the
alleles correspond to HLA binding pockets which engage specific
"anchor" residues of peptide ligands. One HLA molecule will
recognize a range of possible peptides, whereas another HLA
molecule will recognize a different range of peptides. Consequently,
no two individuals will have the same capability of stimulating
an immune response, since they do not bind the same range
of immunogenic peptides. It is estimated that >99% of all
possible peptides derived from foreign antigens are ignored
by any given HLA molecule. Since in the absence of HLA polymorphism
a large number of immunogenic peptides would not be recognized,
the extensive HLA polymorphism in the population reduces the
chance that a given virus or bacterium would not be recognized
by a sizable proportion of the population, reducing the likelihood
of major epidemics or pandemics.
T Cell Recognition Of Antigen Presented By HLA Molecules
Since HLA molecules regulate peptide display to and activation
of the immune system, considerable effort has been devoted
to understanding the molecular basis of peptide-HLA interactions.
These issues are important for defining the biology of T cell
antigen recognition and the properties of a protein that make
it immunogenic or non-immunogenic. Specific antigen recognition
by T cells is dependent on recognition by the T cell receptor
of a three-dimensional complex on the surface of antigen-presenting
cells (APC) comprised of the HLA molecule and its bound peptide.
The peptides are produced by complex antigen processing machineries
within the APC (i.e. proteolytic enzymes, peptide transporters
and molecular chaperones) which generate a pre-selected peptide
pool for association with the HLA molecules. The different
types of T cells require different HLA molecules for antigen
presentation, so-called "HLA restriction" phenomena. T cell
receptors on CD8+ cytotoxic T cells (CTLs) bind peptides presented
by HLA class I molecules, whereas CD4+ T helper cells (Th)
recognize peptides bound to HLA class II molecules. Of the
8-13 amino acid residues of a bound peptide within a class
I or II HLA molecule, only three to four amino acid side chains
are accessible to the T cell receptor, and a similar number
of amino acids are involved in binding to the HLA molecule.
Thymic Education Of T Cells
T cells mature in the thymus to appropriately respond to
foreign pathogens without inadvertently attacking the host.
Under the influence of various thymic resident cells and factors
they elaborate, maturing T cells fall into two categories:
those that are able to discriminate between self and non-self
and can appropriately respond to foreign pathogens without
inadvertently attacking the host, and those which are unable
to appropriately discriminate between self and non-self. Dendritic
cells have been implicated in the deletion, or inhibition,
of T cells reactive to self-antigens, particularly in the
thymus during T cell development or in peripheral lymphoid
organs. The process of self/non-self discrimination by the
maturing T cells is dependent on thymic dendritic cell (DC)
presentation of self-antigens in the context of self-HLA molecules.
When maturing thymic T cells are highly reactive with self-antigen/HLA
complexes, they are deleted so that potentially autoreactive
T cells will not be released into the periphery. If a particular
foreign antigen can be presented in such a way in the thymus
as to fool the maturing T cells into believing that the antigen
is part of self tissue, then T cells capable of reacting with
this antigen will also be eliminated. Indeed, it has been
demonstrated that when mouse thymic DC present transgenically
introduced foreign antigens to developing T cells, the mature
peripheral T cell repertoire of the mouse lacks T cells capable
of reacting with the specific foreign antigen, i.e. it is
tolerant to the foreign antigen. This has raised the possibility
that injection of dendritic cells into an allogeneic recipient
might induce tolerance to a subsequent allograft by causing
deletion or inhibition of alloreactive T cells.
T Cell Recognition Of Alloantigens
Recognition of foreign, or allogeneic, HLA antigens by the
recipient immune system is the major limitation to the survival
of solid organ grafts. The central role of HLA molecules in
allograft rejection is due to their role as restriction elements
for T cell recognition of donor antigens and the extensive
polymorphism displayed by the HLA molecules, which elicit
host immune responses. Although progress has been made in
the short-term survival of transplants, chronic immunologic
rejection remains an impediment to long-term survival.
The primary cause of acute rejection of transplanted organs
is so-called "direct" recognition of whole allogeneic
HLA antigens by receptors on the surface of recipient T cells.
The direct recognition pathway involves recognition by recipient
T cells of donor HLA class I and class II molecules, resulting
in the generation of cytotoxic and helper T lymphocytes which
play a pivotal role in the rejection process. In contrast,
chronic rejection of transplanted organs results from so-called
"indirect recognition" of donor HLA peptides derived
from the allogeneic HLA molecules shed by the donor tissue.
These foreign HLA molecules are taken up and processed by
recipient antigen presenting cells (APC), and peptide fragments
of the allogeneic HLA molecules containing polymorphic amino
acid residues are bound and presented by recipient's (self)
HLA molecules to recipient (self) T cells. Although direct
and indirect recognition of alloantigen generally leads to
adverse graft outcome, tolerance induction may occur following
exposure of the recipient to donor alloantigens prior to transplantation.
Since this strategy is based on the nature and dose of the
antigen as well as the route of administration, understanding
how to control the balance between activation and unresponsiveness
mediated by the direct and/or indirect recognition of alloantigen
is a an area of active research which could lead to development
of new therapies to prolong graft survival.
Indirect allorecognition has been implicated in recurrent
rejection episodes in various transplantation models of cardiac,
kidney and skin grafts. Determinants on donor HLA molecules
can be divided into two main categories: (a) the dominant
allodeterminants that are efficiently processed and presented
to alloreactive T cells during allograft rejection; and (b)
the cryptic allodeterminants that are potentially immunogenic
but do not normally induce alloreactive responses, presumably
due to incomplete processing and/or presentation. Indirect
recognition of allo-HLA peptides is important for the initiation
and spreading of the immune response to other epitopes within
the allograft. So-called "spreading" of indirect
T cell responses to other allo-HLA epitopes expressed by graft
tissue is strongly predictive of recurring episodes of rejection.
Tolerance induction to the dominant donor determinants represents
potential effective strategy for blocking indirect alloresponses
and ensuring long-term graft survival in animal models.
Tolerance Induction
Advances in surgical methods and current immunosuppressive
therapies have led to significant improvement in short-term
graft survival, however long-term survival rates remain poor.
For example, whereas both kidney and heart allografts have
one-year graft survival rates of 85 to 95 percent, only about
50% of transplanted hearts survive five years and only about
50% of kidney grafts survive ten years. Thus, despite being
able to achieve short-term success, these relatively poor
long-term graft survival rates demonstrate the limitations
of the current clinical immunosuppressive regimens to enable
long-term immune evasion by the graft. Consequently, a major
goal of transplantation immunobiologists is to induce donor-specific
tolerance, allowing the long-term survival of human allografts
without the need of HLA-compatibility and without the continuous
recipient immunosupression leading to the concomitant risks
of infection, malignancy, and/or other specific drug side
effects. This would theoretically improve long-term graft
survival, reduce or eliminate the continuing need for expensive,
toxic and non-specific immunosuppressive therapy and enhance
the quality of life.
Insight into some of the mechanisms involved in tolerance
induction has been gained from pre-clinical and clinical studies
in numerous animal models and in patients, particularly those
with liver allografts which typically do not induce a prominent
immune response leading to rejection. One possible mechanism
by which liver transplantation results in allograft tolerance
tolerance may be that the donor or "passenger" lymphoid
cells in the transplanted liver emigrate and take up residence
in the recipient's immune organs, such as the thymus or
lymph nodes. Donor lymphocytes at these sites might "re-educate"
the recipient immune system so that the donor organ is not
recognized as foreign. In an attempt to initiate a similar
process in other organ recipients, transfusions of donor blood
or bone marrow have been used to enhance solid organ graft
survival in animal models and in clinical trials. These studies
are currently ongoing in various organ systems.
Molecular understanding of the cellular immune response
has led to new strategies to induce a state of permanent tolerance
after transplantation. Several approaches have shown promise,
including the use of tolerizing doses of class I HLA-molecules
in various forms for the induction of specific unresponsiveness
to alloantigens, and the use of synthetic peptides corresponding
to HLA class II sequences. Other approaches include alteration
in the balance of cytokines that direct the immune response
away from the TH1 type of inflammatory response and graft
rejection to the TH2 type of response that might lead to improved
graft survival, and the use of agents to induce "co-stimulatory
blockade" of T cell activation. This latter approach
is based on the concept that blockade of a "second signal"
to the T cell enables the signal provided to the T cell receptor
by the HLA-peptide complex to induce antigen specific tolerance.
The experimental use of human dendritic cells as tolerogenic
agents has been limited due to the low frequency of circulating
dendritic cells in peripheral human blood, the limited accessibility
to human lymphoid organs, and the terminal state of differentiation
of circulating human dendritic cells making their further
expansion ex vivo difficult. Dendritic cells are migratory
cells of sparse, but widespread, distribution in both lymphoid
and non-lymphoid tissues. Although the earliest precursors
are ultimately of bone marrow origin, the precise lineage
of dendritic cells is controversial and includes both myeloid-derived
and lymphoid-derived populations. Recent work has revealed
that an expanded population of mature human dendritic cells
can be derived from non-proliferating precursors in vitro
is by culturing bone-marrow derived cells with a combination
of cytokines. This method of enrichment for human dendritic
cells from a precursor population can result in the production
of dendritic cells that are tolerogenic to foreign antigens.
Whether such cells could be useful when co-administered with
an allograft transplant remains to be determined. Nevertheless,
it is clear that considerable progress has been made in the
past few years using approaches to manipulate the immune response
to enable routine donor-specific tolerance, and there is reason
to be optimistic that with better understanding of molecular
and cellular mechanisms this goal could be attained.
3. IMMUNOSUPPRESSIVE AGENTS COMMONLY USED IN ORGAN TRANSPLANT
RECIPIENTS: BENEFITS AND ADVERSE OUTCOMES
Cyclosporine
Cyclosporine has been the single most important factor associated
with improved outcomes after organ transplantation over the
past two decades. CyA binds to a cytosolic cell protein, cyclophilin
(CyP). The CyA-CyP complex then binds to calcineurin and subsequently
blocks interleukin-2 (IL-2) transcription. The binding of
IL-2 to the IL-2 receptors on the surface of T lymphocytes
is a key stimulant in promoting lymphocyte proliferation,
activation, and ultimately allograft rejection. A review of
the first decade of experience with heart transplantation
revealed a total of 379 cardiac allograft recipients worldwide;
actuarial survival rates in this cohort of patients at 1 year
and 5 years were 56% and 31% respectively; the main causes
of death being acute rejection and the side effects of immunosuppression.
With the introduction and widespread use of CyA over the next
decade, survival rates dramatically improved to 85% and 75%
at 1 and 5 years respectively. Similar results were obtained
with other organ transplants, including kidney and lung.
The major adverse effects of CyA are nephrotoxicity, hypertension,
neurotoxocity and hyperlipidemia; less common side effects
include hirsuitism, gingival hyperplasia and liver dysfunction.
CyA nephrotoxicity can manifest as either acute or chronic
renal dysfunction. It is important to note that a number of
drugs commonly used in transplant patients, such as aminoglycosides,
amphotericin B and ketoconazole can potentiate the nephrotoxicity
induced by CyA. More than half the patients receiving CyA
will require treatment for hypertension within the first year
following transplantation. Corticosteroids also potentiate
the side effects of CyA such as hypertension, hyperlipidemia
and hirsuitism. Frequent monitoring of the
serum level is essential to minimize the adverse effects.
One of the major limitations of the original oil-based CyA
formulation (Sandimmune) is its variable and unpredictable
bioavailability. In the mid-90s Neoral was introduced, a new
microemulsion formula of CyA, which has greater bioavailability
and more predictable pharmacokinetics than Sandimmune.
Tacrolimus
Tacrolimus (FK506) is a macrolide antibiotic that inhibits
T-cell activation and proliferation and inhibits production
of other cytokines. The product of Streptomyces tsurubaensis
fermentation, FK 506 was first discovered in 1984 and first
used in clinical studies in 1988 at the University of Pittsburgh.
While the mechanism of action of tacrolimus is similar to
that of CyA, and comparative clinical trials have suggested
similar efficacy, it has been suggested that some groups of
patients may benefit from tacrolimus rather than CyA as primary
immunosuppressive therapy. Unlike CyA, hirsuitism and gingival
hyperplasia occur infrequently with tacrolimus; thus, tacrolimus-based
therapy may improve compliance and quality of life in female
and pediatric transplant recipients. It should be noted that
alopecia has been documented with tacrolimus, but is known
to improve with dose reductions. The decreased incidence of
hypertension and hyperlipidemia with tacrolimus makes it preferable
to CyA in patients with difficult to treat hypertension or
hyperlipidemia. A final indication for tacrolimus has been
as a rescue immunosuppressant in cardiac transplant recipients
on CyA with refractory rejection or intolerance to immunosuppression
(severe side effects). Since tacrolimus is metabolized using
the same cytochrome P450 enzyme system as CyA, drug interactions
are essentially the same. Thus, drugs that induce this system
may increase the metabolism of tacrolimus, thereby decreasing
its blood levels. Conversely, drugs that inhibit the P450
system decrease the metabolism of tacrolimus, thereby increasing
its blood levels. It is important to note that some studies
have indicated a higher incidence of nephrotoxicity with tacrolimus
as compared to CyA.
Azathioprine and Mycophenolate Mofetil (MMF)
Despite being available for more than 35 years, azathioprine
is still a useful agent as an immunosuppressive agent. Following
administration, azathioprine is converted into 6-mercaptopurine,
with subsequent transformation to a series of intracellularly
active metabolites. These inhibit both an early step in de
novo purine synthesis and several steps in the purine
salvage pathway. The net effect is depletion of cellular purine
stores, thus inhibiting DNA and RNA synthesis, the impact
of which is most marked on actively dividing lymphocytes responding
to antigenic stimulation. In currently used immunosuppressive
protocols, azathioprine is used as part of a triple therapy
regimen along with CyA or tacrolimus and prednisone. Mycophenolate
mofetil (MMF), which is rapidly hydrolyzed after ingestion
to mycophenolic acid, is a selective, noncompetitive, reversible
inhibitor of onosine monophosphate dehydrogenase, a key enzyme
in the de novo synthesis of guanine nucleotides. Unlike other
marrow-derived cells and parenchymal cells that use the hypoxanthine-guanine
phosphoribosyl transferase (salvage) pathway, activated lymphocytes
rely predominantly on the de novo pathway for purine synthesis.
This functional selectivity allows lymphocyte proliferation
to be specifically targeted with less anticipated effect on
erythropoiesis and neutrophil production than is seen with
azathioprine.
Early studies in human kidney and heart transplant recipients
showed that MMF, when substituted for azathioprine in standard
triple-therapy regimens, is well tolerated and more efficacious
than azathioprine. In a large, double-blind, randomized multicenter
study comparing MMF versus azathioprine (with CyA and prednisone)
involving 650 patients, the MMF group was associated with
significant reduction in mortality as well as a reduction
in the requirement for rejection treatment. However, there
was noted to be an increase in the incidence of opportunistic
viral infections in the MMF group. The overall greater efficacy
of MMF compared to azathioprine has resulted in MMF generally
replacing azathioprine in triple immunosuppressive protocols
together with steroids and cyclosporine in most solid organ
recipients.
Corticosteroids
Steroids are routinely used in almost all immunosuppressive
protocols after organ transplantation. The metabolic side
effects of steroids are well known and lead to significant
morbidity and mortality in the post-transplant period. Almost
90% of organ recipients continue to receive prednisone at
1-year post-transplant and 70% at three-years post-transplant.
A recent review of over 1800 patients from a combined registry
outlined the morbid complications that patients suffer within
the first year after transplantation. Many of these complications
are known side effects of prednisone, including hypertension
(16%), diabetes mellitus (16%), hyperlipidemia (26%), bone
disease (5%) and cataracts (2%). It is thereby obvious that
avoidance of steroids may decrease morbidity and mortality
after organ transplantation. Two general approaches are used
to institute prednisone-free immunosuppression: early and
late withdrawal.
Withdrawal of prednisone during the first month post-transplant
has resulted in long-term success of steroid withdrawal in
50-80% of patients. In these studies, the use of antilymphocyte
antibody induction therapy appears to increase the likelihood
of steroid withdrawal. Several centers have reported their
results with immunosuppressive regimens that did not include
steroids in the early post-transplant period. Studies reporting
high success rates of 80% have used specific enrolment criteria,
such as excluding patients with recurrent acute rejections
or those with female gender. Review of numerous studies demonstrate
that steroid free maintenance immunosuppression is possible
in atleast 50% of patients, is as safe as triple drug therapy
and may reduce some of the long-term complications of steroids.
Owing to the fact that the majority of acute rejection episodes
occur in the first three months post-transplant, steroid withdrawal
is made after this time period, resulting in long-term success
in about 80% of patients. Generally, there is no need for
conventional induction agents when late withdrawal of steroids
is done.
Anti-Lymphocyte Antibody Therapy
Despite the extensive use of induction therapy using anti-lymphocyte
antibody in solid organ transplantation, their exact role
is unclear. There is no doubt that routine use of these agents
is unwarranted as the generalized immunosuppression induced
by then increased the risk of infections and malignancy. Despite
the lack of consistent data supporting the routine use of
induction therapy with anti-lymphocyte antibody agents, there
is a role in certain select situations. Specifically, patients
with early post-operative renal or hepatic dysfunction may
benefit especially by the avoidance of cyclosporine therapy
while using these induction agents. Anti-lymphocyte antibody
therapy can provide effective immunosuppression for atleast
10 to 14 days without CyA or tacrolimus therapy. It has also
been suggested that patients with overwhelming postoperative
bacterial infections or diabetics with severe postoperative
hyperglycemia may benefit from the comparatively low doses
of corticosteroids required during anti-lymphocyte induction
therapy.
The two main types of induction agents have been either
the polyclonal antilymphocyte or antithymocyte globulins and
more recently the murine monoclonal antibody OKT3. While these
agents have been shown to be effective in terminating acute
allograft rejection and in treating refractory rejection,
the results of comparative studies of outcomes with and without
monoclonal induction therapy have varied, with most studies
demonstrating an effect on rejection that is maintained only
while antibody therapy is ongoing. Without repeated administration,
these agents only delay the time to a first rejection episode
without decreasing the overall frequency or severity of rejection.
More importantly, their use has been associated with an increased
risk of short-term (infections) and long-term (lympho-proliferative
disorders) complications. A complication specific to OKT3
is the development of a "flu-like syndrome" characterized
by fever, chills and mild hypotension, typically seen with
the first dose.
Since antilymphocyte antibodies are produced in nonhuman
species, their use is associated with the phenomenon of sensitization,
leading to decreased effectiveness with repeated use as well
as the possibilty of serum sickness. The development of sensitization
has been linked with an increased risk of acute vascular rejection.
While this association has not been reported by other centers
using OKT3 prophylaxis, it is believed that the development
of immune-complex disease, inadequate immunosuppression due
to decreased OKT3 levels or that OKT3 sensitization may be
a marker for patients at higher risk for humoral rejection
may be responsible for this phenomenon.
Interleukin-2 Receptor Inhibition
A new class of drugs has been developed which targets the
high affinity IL-2 receptor. This receptor is present on nearly
all activated T cells but not on resting T cells. In vivo
activation of the high-affinity IL-2 receptor by IL-2 promoted
the clonal expansion of the activated T cell population. A
variety of rodent monoclonal antibodies directed against the
a chain of the receptor have been used in animals and humans
to achieve selective immunosuppression by targeting only T-cell
clones responding to the allograft. Chimerisation or humanisation
of these monoclonal antibodies resulted in antibodies with
a predominantly human framework that retained the antigen
specificity of the original rodent monoclonal antibodies.
A fully humanized anti-IL2R monoclonal antibody, daclizumab,
and a chimeric anti-IL-2R monoclonal antibody, basiliximab,
have undergone successful phase III trials demonstrating their
efficacy in the immunoprophylaxis of patients undergoing renal
and cardiac transplantation.
Both agents have immunomodulatory effects that are similar
to those of other monoclonal antibody-based therapies (i.e.,
induction of clonal anergy rather than clonal deletion). The
advantages of these agents include their lack of immunogenicity,
long half-lives, ability to repeat dosing, and short-term
safety profile. Daclizumab appears to be an effective adjuvant
immunomodulating agent in cardiac allograft recipients. It
has advantages over conventional induction therapy as it is
more selective and can be used for prolonged and potentially
repeated periods. Studies with larger cohorts are needed to
further study the short-term and long-term survival benefits
for patients following organ transplantation and should determine
the optimal dosing schedules of these new agents.
4. STEM CELL TRANSPLANTATION AND IMMUNO-SUPPRESSION
Materno-Fetal Tolerance
As outlined above, when tissues from an HLA-disparate donor
are transplanted into a recipient they are always recognized
as foreign, and immunosuppression is required to prevent rejection.
An important exception to this is observed in pregnant women
who tolerate their unborn fetus despite the fact that it expresses
a full set of non-maternal HLA antigens inherited from the
father. The mechanisms by which embryonic tissue demonstrates
immune privilege during prenatal development have not yet
been fully elucidated, however it is evident that interactions
between fetus and mother differ substantially from the events
triggered by a classical allograft. Consequently, much work
is being dedicated to the emerging field of materno-fetal
immunobiology in order to enable the development of innovative
strategies to induce tolerance and prevent allogeneic graft
rejection.
When maternal T cells encounter the fetus they demonstrate
adaptive tolerance. In part this may be due to the absence
of expression of MHC class II antigens and low levels of expression
of MHC class I antigens on fetal cells. However, this can
only partly explain the state of prolonged maternal tolerance
since induction of HLA class I and II molecules inevitably
occurs as the fetus matures and differentiates, yet rejection
still does not occur. Consequently, non-fetal aspects of the
placental barrier must be of critical importance in maintaining
prolonged tolerance to the fetus. An important mechanism may
relate to upregulation of the human non-classical HLA class
Ib antigen, designated HLA-G, by the syncytiotrophoblast.
HLA-G molecules bind to inhibitory receptors on natural killer
cells and subsequently protect against maternal rejection
responses. The placenta produces high levels of the anti-inflammatory
cytokine interleukin 10 which stimulates HLA-G synthesis while
concomitantly downregulating MHC class I antigen production,
thus contributing to the tolerance-inducing local environment.
The trophoblast also produces high levels of the enzyme indoleamine
2,3-dioxygenase, which catabolizes tryptophan, an essential
amino acid necessary for rapid T cell proliferation. Annexin
II, found in isolated placental membranes in vitro is present
in placental serum, exerts immunosuppressive properties, and
additionally contributes to fetal allograft survival. Together,
these features indicate that materno-fetal tolerance results
from a combination of transiently reduced antigenicity of
the fetus in combination with a complex tolerance-inducing
milieu at the placental barrier.
Immunogenic Characteristics Of Embryonic And Adult
Stem Cells
Murine and human embryonic stem (ES) cells do not express
HLA class I and II antigens, and demonstrate reduced surface
expression of co-stimulatory molecules important for T cell
activation. Transplantation of murine ES cells demonstrates
long-term graft survival despite the fact that these cells
do acquire HLA class II antigen expression after in vivo differentiation.
Since they are able to accomplish long-term engraftment without
the need for immunosuppression, their inability to induce
an immune response is not likely to be the result of escaping
immune surveillance, but rather due to their ability to colonize
the recipient thymus and induce intrathymic deletion of alloreactive
recipient T cells.
Recently, a population of cells has been described in human
adult bone marrow that has similar functional characteristics
to embryonic stem cells in that they have high self-regenerating
capability and capacity for differentiation into multiple
cell types, including muscle, cartilage, fat, bone, and heart
tissue. While such cells, termed adult mesenchymal stem cells
(MSC), appear to have a more restricted self-renewal capacity
and differentiation potential than ES cells, their functional
characteristics may be sufficient for clinically meaningful
tissue regeneration. A striking recent observation is that
MSC can broadly inhibit T-cell proliferation and activation
by various types of antigenic stimulation, including allogeneic
stimuli. MSCs have been shown to inhibit both naive and memory
T cell responses in a dose-dependent fashion and affect cell
proliferation, cytotoxicity, and the number of interferon
gamma (IFN-gamma)-producing T cells. MSCs appear to inhibit
T cell activation through direct contact, and do not require
other regulatory cellular populations. Similarly to ES cells,
adult bone marrow-derived mesenchymal stem cells (MSCs) do
not express HLA class II molecules, and only low levels of
HLA class I molecules. Despite the fact that MSC can be induced
to express surface HLA class II molecules by in vitro culture
with cytokines such as interferon-gamma, their ability to
inhibit T cell activation results in induction of T cell non-responsiveness
to the MSC themselves, endowing them with potential survival
advantages in the setting of transplantation.
Tolerogenic Effects Of Stem Cell Transplantation
Extending the approaches discussed above using donor-derived
blood transfusions to induce a tolerogenic state to the subsequent
organ, the most promising clinical strategy for tolerance
induction at present is the use of donor-derived hematopoietic
stem cells in conjunction with reduced myeloablative conditioning.
The objective of this therapy is to achieve a state of so-called
mixed chimerism, or the permanent co-existence of donor- and
recipient-derived blood cells comprising all the different
hematopoietic lineages in the same host. This approach has
been tested in a variety of small and large animal settings
and currently available data suggest that stable engraftment
of donor bone marrow reliably renders the host tolerant to
donor antigens and subsequently to any cellular or solid organ
graft of the same donor.
The two underlying mechanisms by which creation of a mixed-chimeric
host results in tolerance induction are (1) thymic deletion
of potentially donor-specific alloreactive T cells, and (2)
nonthymic peripheral mechanisms, such as blocking costimulatory
T cell activation, which facilitate the process of donor bone-marrow
or stem cell engraftment. However, despite the efficacy of
an approach using fully HLA-mismatched stem cells in an allogeneic
host to induce tolerance to a subsequent organ allograft,
the host is placed at a high risk of substantial morbidity
and mortality due to toxicity of the myeloablative conditioning
regimen and potential for graft-versus-host disease, or immune-mediated
attack of the host by the implanted allogeneic stem cells.
In an attempt to overcome these potential limiting toxicities,
investigators have suggested the use of either adult bone
marrow-derived mesenchymal stem cells or preimplantation-derived
embryonic stem (ES) cells for induction of mixed chimerism.
The theoretical advantages of these cell types is their low
level of surface expression of HLA class I and II antigens,
and reduced surface expression of co-stimulatory molecules
important for T cell activation. Rat preimplantation stage
derived embryonic-like stem cells have been shown to successfully
engraft in the recipient bone marrow without the need for
pre-conditioning therapies such as irradiation, cytotoxic
drug regimens or T cell depletion. Long-term partial mixed
chimerism by use of rat preimplantation stage derived embryonic-like
stem cells did not trigger graft-versus-host reactions, in
contrast to the high frequency of this complication in the
clinical setting of allogeneic hematopoietic stem cell transplantation.
Of most interest, the induced partial chimerism enabled the
recipient animals to be tolerant to a subsequent heart allograft.
Allograft acceptance required the presence of an intact thymus,
and rat ES cells were present in the recipient thymus.
Similar results have been reported following transplantation
of human adult bone marrow-derived mesenchymal stem cells
(MSC) into fetal sheep early in gestation, before and after
the expected development of immunologic competence. In this
xenogeneic system, human MSC engrafted, differentiated in
a site-specific manner, and persisted in multiple tissues
for as long as 13 months after transplantation, including
the thymus. Since MSCs do not present alloantigen and do not
require MHC expression to exert their inhibitory effect on
alloimmune reactivity, the possibility exists that they could
theoretically be derived from a donor irrespective of their
HLA type and used to inhibit T-cell responses to transplantation
antigens of an unrelated third party. In initial human clinical
studies, the use human adult bone marrow-derived mesenchymal
stem cells has been shown to successfully enable engraftment
of subsequently infused allogeneic bone marrow in transplant
recipients, reduce the risk of graft-versus-host disease,
and reduce the need for concomitantly administered immunosuppression.
Whether similar results will be obtained when combining adult
bone marrow-derived mesenchymal stem cells with solid organ
allografts remains to be determined, and this is an area of
active research for clinical transplant immunobiologists.
Of broader relevance, if the results relating to long-term
engraftment and survival of adult bone marrow-derived MSC
are confirmed and extended in human clinical studies, they
will have broad implications for the field of tissue and organ
regeneration.
_________________
Selected References
Billingham RE, Brent L, Medawar PB (1953)
Actively acquired tolerance of foreign cells. Nature 172:603-606.
Auchincloss H Jr (2001) In search of the
elusive holy grail: the mechanisms and prospects for achieving
clinical transplantation tolerance. Am J Transplant 1:6-12.
Wekerle T, Kurtz J, Ito H, Ronquillo JV,
Dong V, Zhao G, Shaffer J, Sayegh MH, Sykes M (2000) Allogeneic
bone marrow transplantation with co-stimulatory blockade induces
macrochimerism and tolerance without cytoreductive host treatment.
Nat Med 6:464-469.
Watkins DI (1995) The evolution of major
histocompatibility class I genes in primates. Crit Rev Immunol
15:1-29.
Salter-Cid L, Flajnik MF (1995) Evolution
and developmental regulation of the major histocompatibility
complex. Crit Rev Immunol 15:31-75.
Fowlkes BJ, Ramsdell F (1993) T-cell tolerance.
Curr Opin Immunol 5:873-879.
Marrack P, Kappler J (1990) T cell tolerance.
Semin Immunol 2:45-49.
Van Parijs L, Abbas AK (1998) Homeostasis
and self-tolerance in the immune system: turning lymphocytes
off. Science 280:243-249.
Davies JD, Martin G, Phillips J, Marshall
SE, Cobbold SP, Waldmann H (1996) T cell regulation in adult
transplantation tolerance. J Immunol 157:529-533.
Thomson AW, Lu L (1999) Dendritic cells are
regulators of immune reactivity: implications for transplantation.
Transplantation 68:1-8.
Brent L (1997) Fetally and neonatally induced
immunologic tolerance. In: Brent L (ed) A history of transplantation
immunology. Academic, San Diego.
Matzinger P (1994) Tolerance, danger, and
the extended family. Annu Rev Immunol 12:991-1045.
Janeway CAJ (1992) The immune system evolved
to discriminate infectious nonself from noninfectious self.
Immunol Today 13:11-16.
Mellor AL, Munn DH (2001) Extinguishing maternal
immune responses during pregnancy: implications for immunosuppression.
Semin Immunol 13:213-218.
Fändrich F, Lin X, Chai GX, Schulze M, Ganten
D, Bader M, Holle J, Huang DS, Parwaresch R, Zavazava N, Binas
B (2002) Preimplantation-stage stem cells induce allogeneic
graft tolerance without supplementary host conditioning. Nat
Med 8:171-178.
Liechty KW, et al (2000). Human mesenchymal
stem cells engraft and demonstrate site-specific differentiation
after in utero transplantation in sheep. Nat Med 6(11):1282-6.
Krampera M, et al (2002). Bone marrow mesenchymal
stem cells inhibit the response of naive and memory antigen-specific
T cells to their cognate peptide. Blood, 101(9):3722-3729.
Le Blanc K, Tammik L, Sundberg B, Haynesworth
SE, Ringden O (2003). Mesenchymal stem cells inhibit and stimulate
mixed lymphocyte cultures and mitogenic responses independently
of the major histocompatibility complex.Scand J Immunol. 57(1):11-20.
|