Transplantation and Rejection

Published on 18/02/2015 by admin

Filed under Allergy and Immunology

Last modified 22/04/2025

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 3185 times

Chapter 21 Transplantation and Rejection

Summary

Transplantation is the only form of treatment for most end-stage organ failure.

The barrier to transplantation is the genetic disparity between donor and recipient.

The immune response in transplantation depends on a variety of factors. Host versus graft responses cause transplant rejection. Histocompatibility antigens are the targets for rejection. Minor antigens can be targets of rejection even when donor and recipient MHC are identical. Graft versus host reactions result when donor lymphocytes attack the graft recipient.

Rejection results from a variety of different immune effector mechanisms. Hyperacute rejection is immediate and caused by antibody. Acute rejection occurs days to weeks after transplantation. Chronic rejection is seen months or years after transplantation.

HLA matching is one of two major methods for preventing rejection of allografts. The better the HLA matching of donor and recipient, the less the strength of rejection.

Successful organ transplantation depends on the use of immunosuppressive drugs. 6-MP, azathioprine, and MPA are antiproliferative drugs. Ciclosporin, tacrolimus, and sirolimus are inhibitors of T cell activation. Corticosteroids are anti-inflammatory drugs used for transplant immunosuppression. Antibodies to the IL2 receptor, or to leukocytes, are widely used.

The ultimate goal in transplantation is to induce donor-specific tolerance. There is evidence for the induction of tolerance in humans and novel methods for inducing tolerance are being developed. Alloreactive cells can be made anergic. Immune privilege can be a property of the tissue or site of transplant.

Shortage of donor organs and chronic rejection limit the success of transplantation. Living donation is one way to overcome the shortage of donor organs. Alternative approaches are being investigated. The favored animal for xenotransplantation is the pig.

Transplantation is the only form of treatment for most end-stage organ failure, and it is a central topic for immunologists for two reasons:

As a clinical procedure, transplantation is used to replace tissues or organs that have failed. The first successful transplants were those of the cornea, first described in 1906.

World War II provided an important impetus, with the problems of skin grafting airmen who had extensive burns motivating a number of scientists, most notably Peter Medawar, to investigate the immunological basis of graft rejection.

The subsequent demonstration by the Medawar group that it was possible to manipulate a recipient animal so that it accepted grafts from an unrelated donor animal encouraged the subsequent clinical development of transplantation. The discovery (by Calne and others) of immunosuppressive drugs and agents then allowed the widespread practice of transplantation in the last three to four decades of the 20th century.

In modern practice many transplants are performed routinely (Fig. 21.1). In addition to routine transplantation of the cornea, kidney, heart, lungs, and liver there is increasing interest in transplanting other organs, such as whole pancreas or islet cells for diabetes mellitus and also small bowel.

In general most transplants use organs from dead donors (cadaveric transplants), though there is an increasing number of living donors (usually related to the recipients) for kidney transplantation (see below).

Hematopoietic stem cell transplantation

Hematopoietic stem cell transplants are performed for two main reasons. One is to treat children who have inherited immune deficiencies. These children are very prone to infection, and will normally die young as a consequence. However, if they are given stem cells from a healthy donor, the infused stem cells can replace the defective bone marrow stem cells. The stem cells can then mature and into fully effective immune cells, thus giving the child a functioning immune system.

The second major application is for patients with leukemia. It is possible to eradicate the patient’s leukemic cells with chemotherapy and radiotherapy. However, this also results in destruction of the patient’s stem cells in the bone marrow and circulation. The patient therefore becomes immunodeficient and will die of infection. Stem cell transplantation can ‘rescue’ the patient by providing a fresh source of stem cells. In some cases the stem cells are autologous (in which they are harvested before chemotherapy, stored, and then infused back into patients after the therapy is over). In these settings there is no risk of graft versus host disease (see below). However, there is a risk that leukemic cells will be present in the stored stem cells, and will then grow in the patient. In other cases the stem cells come from a well matched donor. This removes the chance of carry over of leukemic cells, but does run the risk of graft verus host disease. In some forms of leukemia it has been shown that there is a graft versus leukemic effect, in which the allogeneic T cells mount a response against any leukemic cells remaining in the patient and prevent them from growing.image

Organ donation

The most limiting factor for organ transplantation is the shortage of donors. There are many more people who would benefit from an organ than there are organs available. Indeed, given the good survival of most forms of transplantation, there are some conditions where it would be beneficial to receive a transplant but they are never considered and treated in other ways because there will always be higher priority cases.

There are several solutions to this issue. One is to increase the number of donors, using advertising and donor recruitment campaigns. This also involves raising the awareness in the public about organ donation, and this can be a particular issue in countries where there are cultural or religious barriers to donation. One possibility in this area is legislation; some countries operate a ‘presumed consent’ policy whereby everyone is assumed to have given consent for their organs to be used, unless they have indicated otherwise. Other countries operate an ‘opt in’ approach where individuals (or their relatives) have to give permission for organs to be used. It is also important to improve the donation rate in ethnic minority groups, who often have a low donation rate. The second is to improve the process by which relatives are approached about organ donation, and to ensure that there are enough facilities to allow all the organs to be harvested (remembering that one donor can provide many organs for transplantation). Transplant coordinators (as they are termed in the UK, but equivalent positions are found in other countries) are key to this process, as are the surgical teams that obtain the organs. The third is to relax the donor selection criteria, to use organs that would not have been used previously. Thus the use of non heart beating donors is increasing, and there is increased use of ‘marginal donors,’ for example from older donors or those that have diseases that would have previously excluded them.

The final ways to increase donation is the use of living donor or using animals as donors (xenotransplantation), both of which are discussed below.

Ethical issues are an important factor in living donation

The shortage of organs from cadaveric donors has increased the use of living donors. Clearly this is only possible in situations where donation of an organ poses little risk to the living donor. Kidney transplantation is routinely performed from living donors, as it is quite possible to live with just one healthy kidney. It is also possible to donate a lobe of a liver, as it can grow back again. Donation of a lung lobe is also possible, though this is a much riskier procedure. Hematopoietic stem cell transplants also involve living donors, though the issues with this form of transplant are somewhat different than for solid organ transplants as the stem cells are rapidly replaced.

Before any donation from a living donor is carried out, it is important to ensure that the potential donor is healthy, that the donation is compatible (of the appropriate blood group), and that the donor is aware of the risks and has not been coerced into giving an organ. It would be unethical for a donor to be put under extreme pressure to donate an organ, and the transplant teams must take great care to ensure that this is not happening. In the majority of cases donation is carried out between close relatives (including spouses) or sometimes friends. However, there are a few ‘altruistic donors’ that give their organs to be used by whoever needs them. In most countries it is not allowed to sell organs for transplantation, and the majority of those involved in transplantation would see that as unethical. However, there is a real problem with ‘transplant tourism’ where people from rich, developed, countries travel to buy organs from poor, marginalized, donors.

Organ exchange programs have been set up to maximize the number of transplants performed. In the simplest cases these are paired donations. Jane Smith may need a kidney and have a relative, Steve Smith, who is willing to donate it. However, if Steve Smith’s kidney is incompatible with Jane then he cannot donate it to her. However, if there is another couple in a similar predicament, so that Jim Jones needs a kidney and Stella Jones is willing to donate one but they are incompatible, then it might be possible for Stella Jones to donate her kidney to Jane Smith and for Steve Smith to donate to Jim Jones. More complex arrangements are also possible, with three or more pairs donating in such a way as to maximize the number of transplants. Indeed it is possible to use an altruistic donor to initiate a chain of donation, such that they donate to recipient A, recipient A’s potential donor gives to recipient B, their donor gives to C and so on, until the final recipient is an individual who does not have a suitable donor.

In addition to transplantation of organs, there is a large program of transplanting hematopoietic stem cells (cells capable of regenerating blood cells), for example in patients with leukemia or with primary immune deficiencies. These stem cells were previously normally harvested from the bone marrow of (living) donors, though increasingly peripheral stem cells obtained from the blood are used – stem cell transplantation has its own particular problems.

Clinical trials of stem cell therapy, using mesenchymal stem cells or embryonic stem cells are now underway. The aim of these is that the stem cells can differentiate and repair damaged organs. In some cases the stem cells are taken from the patient, in which case there is no immunological issues. However, if they are taken from another individual then rejection is possible.

Genetic barriers to transplantation

The main immunological problem with transplantation is that the grafted organ or tissue is seen by the immune system as ‘foreign’ and is recognized and attacked – leading to rejection of the organ.

Transplantation is normally performed between individuals of the same species who are not genetically identical, and the antigenic differences are known as allogeneic differences, and result in an allospecific immune response (Fig. 21.2).

However, it is also possible in experimental circumstances (and possibly in the future in the clinical setting) to perform grafting between different species. This is termed xenotransplantation, and the antigenic differences between donor and recipient form the xenogeneic barrier.

Transplantation can also be performed within an individual (e.g. skin grafting), when it is known as an autograft.

Syngeneic or isografts can be performed between genetically identical individuals. This can occur clinically for identical twins, but is more commonly seen in experimental settings with inbred strains of animals.

In the case of autografts and isografts there should be no antigenic differences between donor and recipient, and so no immune response. This can be readily illustrated using transplantation of skin or organs between inbred strains of animals (Fig. 21.3).

Graft rejection

There is a high frequency of T cells recognizing the graft

One of the main features of the immune response against a transplanted organ is that it is much more vigorous and strong than the response against a pathogen, such as a virus. This is largely reflected by the frequency of T cells that recognize the graft as foreign and react against it.

Thus, in a naive or unimmunized individual fewer than 1/100 000 T cells respond upon exposure to a virus or a protein immunization; however, 1/100–1/1000 T cells respond to allogeneic antigen-presenting cells (APCs). This is reflected in the strong T cell response (proliferation) seen when naive T cells are stimulated with allogeneic dendritic cells (Fig. 21.4)

Why is there such a high frequency of allospecific T cells?

Several models seek to explain why there is such a high frequency of allospecific cells in the T cell repertoire.

The first model (high determinant density model) (Fig. 21.w1) suggests that allospecific T cells recognize the foreign MHC molecules directly, with a low affinity, in a peptide-independent manner. The affinity of the interaction would normally be too low to activate the T cells; however, because the T cells see the MHC molecules directly, and are not recognizing the peptide, this low affinity is compensated for by the high concentration of MHC molecules on allogeneic cells.

The second model (multiple determinant model) states that what the allospecific T cells are recognizing are peptides derived from normal, non-polymorphic, host proteins that bind to and are presented by the foreign MHC molecules, but are not presented by self MHC. Due to lack of presentation by self MHC, the T cell repertoire is not tolerant to such peptides. The high frequency of the response is due to the large number of such antigens that can be presented by the graft.

Indirect recognition is important in chronic rejection

In a primary alloresponse (see Fig. 21.4), most of the alloreactive CD4 or CD8 T cells directly recognize the donor MHC molecules.

However, there are other forms of alloresponse, including:

The indirect response is very similar to conventional T cell recognition of normal antigens, such as those from a pathogen, which are processed by host APCs and presented in the context of host MHC molecules.

Nevertheless, the indirect pathway of recognition is important during chronic rejection, when the number of donor-derived professional APCs is no longer high enough to stimulate a direct immune response. It is also important in the rejection of corneal grafts because the cornea lacks large numbers of APCs.

Minor antigens can be targets of rejection even when donor and recipient MHC are identical

Although the MHC is the major target of the alloimmune response, there are also minor histocompatibility antigens. These can serve as targets of rejection even when the MHC is identical between donor and recipient.

The nature of most minor histocompatibility antigens is unknown, though they are assumed to be normal polymorphic molecules, peptides from which bind to host MHC and induce an immune response. In some cases they are expressed in a tissue-specific manner.

Perhaps the best studied minor histocompatibility antigen system is the H-Y system. These are antigens encoded by the Y chromosome, and so are expressed only on male cells. Thus, following immunization, it is possible to demonstrate immune responses and rejection of male organs or skin following transplantation mediated by female animals (2X chromosomes) against male cells (X and Y chromosome). It is not possible to show responses against female antigens by male animals because the male animals have one X chromosome, and so are tolerant to all antigens encoded on it (Fig. 21.6).

Graft versus host reactions result when donor lymphocytes attack the graft recipient

Although it is usual to think of the immune response recognizing and destroying the transplanted organ, the situation is different when competent immune cells are transplanted into a recipient. This can happen during bone marrow transplantation, when normal donor T cells may be infused into the recipient. In such circumstances the T cells can recognize the MHC molecules and/or minor histocompatibility antigens of the recipient as foreign, and produce an immune response against the recipient. This is known as graft versus host disease (GvHD).

GvHD can be lethal, causing damage in particular to the skin and gut. It can be demonstrated in animal models by transfer of bone marrow to irradiated recipient animals (Fig. 21.7). It can be avoided by:

Immune effector mechanisms in graft rejection

Rejection of organs or tissues can occur at various times, each of which is associated with different immune effector mechanisms (Fig. 21.8). These are:

Hyperacute rejection is immediate and mediated by antibody

Hyperacute rejection is seen when the recipient animal has pre-existing antibodies that are reactive with the donor tissue. This may be because:

A special case is seen in xenotransplantation, where humans and Old World monkeys and apes all have pre-existing antibodies to a carbohydrate antigen α-galactosyl. This carbohydrate is expressed on cell surface proteins of all other donors. Therefore, a xenotransplant of a cellular organ from a pig (or most other species) into a primate is at risk of hyperacute rejection.

Hyperacute rejection is seen within minutes of connecting the circulation into the transplanted organ. It is caused by the pre-existing antibodies binding to the endothelial cells lining the blood vessels and by initiating immune effector functions.

Complement activation can lead to death of the endothelium, or, when the damage is sub-lethal, activation of the endothelial cells. This not only causes an inflammatory response, increasing vascular leakage, but can also cause blood coagulation The result is rapid destruction of the graft (Fig. 21.9).

Prevention of hyperacute rejection is performed by carefully avoiding transplanting an organ into an individual with pre-existing antibodies to that tissue. This is done by:

This involves incubating donor leukocytes with recipient serum in the presence of complement; cell death indicates the presence of anti-donor antibody and is a contraindication to proceeding with transplantation. Such cross-matching is normally performed immediately before surgery. There is increasing success in transplanting across ABO incompatible barriers, though this requires very careful preparation of the recipient.image

ABO incompatible transplantation

As discussed above, transplantation across an ABO incompatibility barrier results in hyperacute rejection of the organ. However, there is increasing experience in such transplants. To give a practical example, let us take a patient Isaac Bloggs who has blood group B and requires a kidney. He therefore has anti-A antibodies. His brother, Mohammed Bloggs, is willing to donate a kidney and is a perfect HLA match. However, he is blood group A, and so antibodies in Isaac’s blood will bind to Mohammed’s cells and destroy them though hyperacute rejection. However, the transplant team decide to go ahead with the transplant.

The first stage is to deplete Isaac’s blood of anti-A antibodies. This can be done by plasmapheresing him (taking his blood out, replacing the plasma that contains the antibodies and then returning it to him). Alternatively his blood can be passed down an affinity column that removes the antibodies. This process will need to be repeated multiple times in order to get the levels of anti-A antibody low. In some hospitals Isaac will also be given antibodies to deplete B cells or have his spleen removed, to reduce the immune response following transplantation.

Once the antibody levels are low enough, then Mohammed’s kidney can be transplanted into Isaac. There is a need for careful monitoring in the early days, and it may be necessary to continue plasmapheresis. Isaac will also receive the conventional immunosuppressive drugs.

In most cases these transplants do very well. Interestingly the levels of anti-A antibody in Isaac are likely to return to close to the levels seen before the procedures over a few months. However, these antibodies do not destroy the donated graft, even though they would have resulted in hyperacute rejection if they had been there at the time of transplantation. The reason is that the cells in the graft, most probably the endothelial cells, have undergone a process termed accommodation, in which they survive even though there are high levels of antibody capable of binding to them. The process of accommodation involves upregulation of a number of protective genes in the cells of the donated kidney.

In the case of xenotransplantation, one solution being developed is the generation of pigs that lack the α-galactosyl carbohydrate epitopes. This is done by knocking out the gene encoding the enzyme (α-galactosyl transferase) that generates this carbohydrate. In preclinical models in which pig organs are transplanted into primates, this approach has led to prolonged graft survival with no evidence of hyperacute rejection.

Acute rejection occurs days to weeks after transplantation

Acute rejection is normally seen days to weeks after transplantation, and is caused by activation of allospecific T cells capable of damaging the graft.

Donor dendritic cells (sometimes called passenger leukocytes) play an important role in triggering acute rejection. Dendritic cells that are present in the organ, following transplantation into the recipient, migrate to the lymph nodes draining the organ and stimulate a primary alloimmune response.

The importance of these dendritic cells can be shown by ‘parking experiments’ in which:

However, if the third animal is injected with donor-derived dendritic cells there is rapid graft rejection. These data highlight the contribution of donor dendritic cells in initiating the alloresponse.

Although the direct pathway is thought to predominate in acute rejection, the indirect alloresponse, though significantly weaker, can also cause acute rejection in some animal models.

Once activated the T cells migrate to the organ and lead to tissue damage by standard immunological effector mechanisms (Fig. 21.11). These include:

If the animal or patient has already been exposed to the alloantigens expressed by the graft, and as a consequence has been immunized, there will be alloreactive memory cells. This will lead to a much more rapid (accelerated) rejection of the graft (Fig. 21.12).

Chronic rejection is seen months or years after transplantation

In vascularized organs chronic rejection presents as occlusion of blood vessels, which on histological analysis show a thickening of the intima, similar in some respects to the thickening seen as a result of atherosclerosis (Fig. 21.13). Smooth muscle cell proliferation is often seen, together with a macrophage infiltrate (together with some lymphocytes). This eventually leads to blockage of the blood vessels and subsequent ischemia of the organ.

A number of mechanisms can lead to chronic rejection. They include:

Non-immunological processes are also important, such as:

In some cases, initiation of chronic rejection may be immunological in nature, but its progression is due to non-immunological mechanisms.

Chronic rejection responds poorly to current immunosuppressive therapy. Therefore, although there has been considerable improvement in overall graft survival over the past decades, this improvement is mostly seen in the first year following transplantation – the subsequent survival of grafts has hardly altered over the past 20–30 years (Fig. 21.14). This indicates the need to improve the treatment of chronic rejection.

HLA matching is important to prevent rejection

The two major methods for preventing rejection of allografts are:

However, in animal models (and hopefully in the clinic) there are techniques that can induce tolerance to an organ such that the immune system of the recipient ‘learns’ to treat the donor organ as ‘self’ and not destroy it. As discussed below, the ability to induce donor-specific tolerance is the ‘Holy Grail’ of transplantation immunology.

The better the HLA matching of donor and recipient, the less the strength of rejection

The major antigenic differences recognized by the alloimmune response are found on the MHC molecules (HLA in humans). These highly polymorphic molecules have a vital role in presenting antigens to T cells.

There are many different alleles of the MHC molecules, and one way to reduce the strength of a rejection response is to match the donor and recipient so that they share as many alleles as possible. In general matching is now performed using molecular techniques, with polymerase chain reactions (PCR) that are specific for the different alleles.

In humans HLA matching is rarely perfect between unrelated donors because of the difficulty in matching all MHC class I and class II gene loci and the high level of polymorphism at each locus.

The extensive polymorphism means that there can hundreds of variants of each antigen. For example the number of (protein) variants of the HLA molecules identified up to April 2011 was: HLA-A 1176; HLA-B 1641; HLA-C 808; HLA-DRA 2, -DRB 774; HLA-DQA 27, -DQB 106; HLA-DPA 16, -DPB 129.

Therefore, even before one considers other polymorphic molecules associated with the HLA locus, there is considerable complexity, which makes it highly unlikely for there to be a complete match.

In cases where the transplant is between living related donors (such as brothers and sisters) there is a greater opportunity for a match because in general the HLA locus is inherited en bloc as a single set (or haplotype) from each parent.

Loci outside the MHC can also lead to rejection (the minor histocompatibility antigens). However, there is no attempt to match for these antigens because there is little possibility of getting a good match and the effect of matching any single minor antigen is too small to be significant.

It should be noted that even when siblings are perfectly matched at the MHC locus they will not be matched (unless they are identical twins) for the minor histocompatibility antigens.

Immunosuppressive drugs

The success of organ transplantation is entirely dependent on the use of immunosuppressive drugs that control the alloimmune response. Although rejection episodes still occur, they are usually kept in check by the drugs so that lasting damage is minimized.

Over recent decades there has been a marked improvement in short-term success rates, such that over 90% of kidney transplants are functioning 1 year after transplantation. The major reason for these improved success rates is the advent of more powerful immunosuppressive agents.

Despite the continuing interest in strategies to promote specific immunological tolerance, clinical transplantation is likely to require non-specific immunosuppression for some years to come. The present challenge is to use the currently available agents intelligently to minimize side effects while preserving graft function.

The commonest cocktail of drugs used for kidney transplant patients involves three agents, each of which has a distinct mode of action:

Usually three agents are used in the early post-transplant period while the anti-donor immune response is at its peak. Increasingly monoclonal antibodies are also being used for preventing rejection.

Numerous clinical trials are addressing the safety of withdrawing one of these three agents within weeks or months of transplantation. It appears that maintenance immunosuppression with two drugs is safe and has an improved side effect profile.image

Ciclosporin, tacrolimus, and sirolimus are inhibitors of T cell activation

Ciclosporin inhibits the production of IL-2

Drugs that inhibit T cell activation are the mainstay of immunosuppressive regimens. The first such drug to be discovered was ciclosporin, a fungal metabolite (Fig. 21.w2).

The introduction of ciclosporin revolutionized clinical transplantation in that it led to a marked improvement in early success rates, which rose from approximately 70% to over 90%. Furthermore, the doses of other drugs that were required were substantially lower, so drug side effects were less troublesome.

Because of the much greater potency of ciclosporin, and its ability to prevent early acute rejection, transplantation of organs other than kidney became routine. Programs of heart, liver, and more recently lung transplantation were established in major transplant centers around the world.

The key effect of ciclosporin is to inhibit the production of the major growth factor for T cells, interleukin-2 (IL-2).

The intracellular mechanism of action of ciclosporin involves binding to cyclophilin, and the consequent inhibition of the calcium-dependent phosphatase, calcineurin, which would otherwise activate the NFAT complex, and lead to IL-2 gene transcription.

Corticosteroids are anti-inflammatory drugs used for transplant immunosuppression

Corticosteroids, given in combination with azathioprine, were the mainstay of immunosuppression for several decades. Only now is the possibility of rapid corticosteroid withdrawal, after 1 or 2 weeks, being explored.

Corticosteroids are pharmacological derivatives of the glucocorticoid family of steroid hormones, and act through intracellular receptors that are almost ubiquitously expressed.

The anti-inflammatory effects of steroids are highly complex, reflecting the fact that as many as 1% of genes may be regulated by glucocorticoids. Some of the most important effects are:

The problem of side effects is a major issue with the use of corticosteroids, particularly at high doses. Central weight gain, fluid retention, diabetes mellitus, bone mineral loss, and thinning of the skin, all result from protracted corticosteroid use. It is for this reason that much attention is being devoted to protocols that allow steroid minimization. However, for corneal transplants, where topical (local) administration of the drug is possible (thus minimizing side effects), corticosteroids are the main form of immunosuppression used.

Antibody therapy

Antibodies have long been used for preventing graft rejection. One of the earliest forms of immunosuppression was anti-lymphocyte globulin or anti-thymocyte globulin; polyclonal antibodies raised by immunizing rabbits or horses with lymphocytes or thymocytes. These antibodies deplete circulating T cells. These are still used, though they are increasingly being replaced with monoclonal antibodies. The first monoclonal antibody licensed for any purpose was Muromonab-CD3 which was approved by the FDA in 1986 for the treatment of rejection episodes following renal transplantation. This binds to CD3 on T cells, and suppresses T cell responses. The action of Muromonab binding CD3 can result in a cytokine release syndrome on infusion, caused by the secretion of inflammatory cytokines by the T cells following engagment of CD3. Two anti IL2 receptors (daclizumab and basiliximab) are now widely used in transplantation, as part of standard immunosuppressive agents. These antibodies operate by blocking the IL2 receptor, rather than depleting T cells. Alemtuzumab (commonly known as CAMPATH-1 H), which depletes CD52 expressing leukocytes, has been widely used for transplantation though it is not yet licensed for that application. Other antibodies are being investigated for particular applications in transplantation (for example the depletion of B cells). Antibody like molecules (e.g. abatacept and belatacept which are two forms of a fusion protein of CTLA4 with the Fc portion of an antibody (see Chapter 8)) are also being evaluated for clinical use.

Induction of donor-specific tolerance

Although generalized immunosuppression has been highly successful in preventing graft rejection, it comes at a price. This includes:

It would therefore be desirable to induce tolerance to the graft whereby the immune system specifically becomes non-responsive to the donor antigens, yet is still capable of responding normally to other antigens.

Tolerance to grafts was first demonstrated by Peter Medawar’s group. They showed that, if allogeneic cells were injected into a neonatal animal, when the animal became adult it would be tolerant to tissue from the donor and would accept grafts without the need for immunosuppression. There have been numerous examples of inducing tolerance to grafts in animal models since, but it has been difficult to translate this into the clinical setting.

One of the difficulties in the clinical setting is to demonstrate that tolerance really exists. In an animal model it is relatively easy to demonstrate tolerance by:

However, this is more difficult in humans.

There is evidence for the induction of tolerance in humans

There are two sources of evidence for the induction of tolerance in humans.

First there are patients who have received grafts, but are no longer on immunosuppressive regimens because they cannot tolerate the drugs. They can show long-term graft survival. This is not formal evidence of tolerance, but it is highly suggestive that some people can have an operational tolerance whereby they fail to destroy their organ graft.

Second, it is possible to look at the frequency of alloreactive T cells in patients with grafts. In some groups there is a reduced frequency of these cells, but the response to other antigens remains normal (Fig. 21.15). Again, this is not a formal proof of tolerance because it is not yet known how the in-vitro assays relate to the response in patients. However, it does indicate that it might be possible to develop tests that will allow us to monitor the development of tolerance in patients, and so know how to tailor treatment to the individual (e.g. removing them from immunosuppression when indicated).

Third, there are now studies that do demonstrate long term graft survival (and so apparent tolerance) in patients that have received joint renal and hematopoetic stem cell transplants. These patients had a short course of immunosupression, and have since seen long term graft survival with no rejection. While the number of patients treated is limited, these data are very encouraging.

Alloreactive cells can be made anergic

In the peripheral organs tolerance induction can result from deletion. However, it is also possible for alloreactive cells to be anergized. Anergy describes a state in which the cell is not deleted, but has been rendered unresponsive to further stimulation by the same antigen.

Blockade of co-stimulatory molecules such as CD80 and CD86 with agents like CTLA-4–Ig (a fusion protein between CTLA-4, a ligand for CD80 and CD86, and the Fc part of an antibody molecule) can be used to induce anergy in alloreactive cells (Fig. 21.16). However, it should be noted that the situation can be more complex than this. In many APCs cross-linking of CD80 and CD86 with CTLA-4–Ig results in upregulation of an immunomodulatory enzyme indoleamine 2,3-dioxygenase (IDO). This enzyme catabolizes tryptophan, and as a result prevents T cell activation both as a result of:

The role of IDO in immune regulation was first recognized in the placenta, where it protects the fetus from immunological rejection – inhibition of IDO causes rejection of histoincompatible fetuses.

Another alternative is to induce a regulatory response to the alloantigen. The phenomenon of T cell regulation has long been recognized (Fig. 21.17), and can be shown in experimental models by transferring T cells from a tolerant animal to a naive recipient, and showing that this results in a transfer of the tolerance. Several types of T cell are capable of regulating the immune response, and strategies that seek to expand these cells may be one method to induce tolerance in vivo.image

Immune privilege can be a property of the tissue or site of transplant

Although a failure to reject a graft could be the result of tolerance (or immunosuppression), an alternative is that the graft is immune privileged and protected from the immune response against it (see Chapter 12):

It is important to note that immune privilege is not an absolute term. Corneal grafts are rejected, albeit less vigorously than other grafts. It is probably best to think of immune privilege as a spectrum ranging from grafts that show a high degree of privilege (cornea) to those where the immune response against them is very strong (skin), with other grafts somewhere in between.

Several mechanisms can be responsible for immune privilege. These include ‘ignorance’ – the immune system does not see the graft. The cornea is not vascularized, and has a poor lymphatic drainage. It also has a very low concentration of dendritic cells in the center. These features reduce the ability of the graft to stimulate an immune response against it. If this ignorance is disrupted (e.g. by inducing vascularization in the graft bed) then the cornea is more rapidly rejected.

In addition the tissue can deviate the immune response. In the anterior chamber of the eye there is a cytokine environment (high in TGFβ and α-melanocyte stimulating hormone) that deviates the immune response away from a tissue destructive to a non-inflammatory response.

Finally, the tissue being transplanted can itself be privileged. This is also seen in the cornea, which expresses high levels of Fas ligand.

Limitations on transplantation

Two major issues limit success of transplantation:

The second of these problems, chronic rejection, would be solved if we were able to induce tolerance to grafts in patients. To do this it is necessary that:

These assays are still under development but may involve measuring the frequency of alloreactive and/or regulatory T cells in patients, as well as other immunological biomarkers. At present it is not clear which tolerance induction procedure is most likely to work in clinical transplantation, and further work in primate models is needed to address this issue.

Xenotransplantation

The most favored animal for developing xenotransplantation is the pig, for reasons that include the physiological and anatomical compatibility between pigs and humans and the ability to breed large numbers of animals rapidly.

There are several barriers to xenotransplantation, including public acceptability, safety, and scientific issues.

The main safety concern is the risk of transmission of viruses from the pig to the human, though this is a diminishing concern. It should be noted that acellular xenografts have been performed for many years and are highly successful (e.g. in the case of pig heart valves, which can be used to replace diseased valves). These are not subject to immunological rejection, nor are they potential sources of viral infection.

The scientific issues revolve around preventing graft rejection. As indicated above, a pig organ transplanted into a human would undergo hyperacute rejection as a result of preformed anti-α-galactosyl antibodies in the recipient’s circulation. One solution has been to engineer pigs (by nuclear cloning from cell lines) that lack the galactosyl transferase enzyme responsible for creating α-galactosyl residues. Organs from these pigs show reasonably long-term survival in primates.

As well as offering scientific challenges, there are also opportunities for using pig organs. The ability to genetically engineer the pig should make it easier to develop tolerance to the organ. This, as well as the ability to carry out transplants in a pre-planned manner, offers considerable advantages over conventional transplantation.

Critical thinking: Kidney transplantation (see p. 440 for explanations)

Mrs X has diabetes mellitus, and this caused severe damage to her kidneys. This complication is called diabetic nephropathy, and is one of the major indications for kidney transplantation. Mrs X was on dialysis treatment, but this was not working well for her and she was advised that she would benefit from renal transplantation. However, it proved very difficult to find a suitable cadaveric donor for Mrs X and it was suggested that a family member might donate an organ. All her immediate family – her husband, five children, and two brothers – agreed to be considered as donors.

The HLA types and blood groups of the family members are shown in the table. On the basis of these tests a donor was selected and the transplant was performed. Despite successful surgery, the kidney soon turned dark and swelled. This started to happen within a few minutes of the restoration of blood flow through the transplant, and necessitated the immediate removal of the graft.

Four years later Mrs X was still very ill on dialysis, no cadaveric donor was available, and it was decided to try again with a living related transplant. Another member of the family was selected to donate a kidney and it functioned well from the onset. Mrs X was given triple immunosuppression. She had only one rejection episode at about 3 weeks after grafting, and this was treated successfully with anti-rejection therapy. There were no other problems.

image

The kidney continued to work for 8 years, but its function gradually declined from the fourth year onwards. It seemed there was little the doctors could do to prevent this worsening situation, and Mrs X eventually had to return to dialysis.

1. What are the difficulties in finding a donor organ?

2. Comment on the HLA relationships between Mrs X and her brothers.

3. Comment on the relationships between the children of Mrs X.

4. Classify each member of the family in terms of their HLA relationship to Mrs X (HLA identical, HLA haplotype match, complete HLA mismatch).

5. In terms of HLA matching alone, who was the best donor for Mrs X?

6. Consider what effect the blood group antigens had on the choice of donor. From whom could kidneys have been transplanted, and who would not have been suitable?

7. Of those who had a compatible blood group, who would you have chosen as the best donor? Explain your reasoning.

8. The outcome of the transplantation was a disaster! By what mechanism was the graft attacked?

9. Why was Mrs X at a greater risk of this untoward reaction?

10. What laboratory tests are used to avoid this rejection reaction, and what seems to have gone wrong on this occasion?

11. Four years after the first transplant it was decided to try again with a living related donor. Of all the family members, who would you have chosen as the donor and whose kidney was most likely to survive in Mrs X?

12. What is triple therapy immunosuppression?

13. What type of rejection occurred at 3 weeks after transplantation, and what immunological mechanisms were involved?

14. What is anti-rejection therapy?

15. There were no other problems with Mrs X. Can you think of some of the problems that might arise in a transplant recipient?

16. Why did the function of the transplant gradually decline, and why could the doctors not stop this process?