Advances in Immunosuppressive Therapy



Fig. 45.1
Direct and indirect pathways of allorecognition. Signal 1 is delivered by a peptide-HLA complex. Signal 2, also known as costimulatory sign, is delivered by an array of cell surface molecules on the T-helper cell and the antigen-presenting cell (APC). DAPC donor APC, RAPC recipient APC, TH T-helper lymphocyte, TC cytotoxic T lymphocytes



As outlined before, graft rejection is due to a complex interaction of different parts of the immune system, including B and T lymphocytes, APCs, and cytokines. Interleukin 2 (IL-2) is secreted by activated CD4+ T-helper lymphocytes and acts as a pivotal, early signal in immune activation. Among its functions, IL-2 engenders increased T-cell responsiveness, clonal expansion of alloreactive T lymphocytes, B lymphocytes, and acquisition of the cytolytic phenotype by host T lymphocytes. Cellular-mediated rejection is the result of an attack of activated T cells on the transplanted organ. Activated B lymphocytes further differentiate into plasma cells and producing antigen-specific antibodies. Antibody-mediated rejection is the result of binding of these antibodies on the transplant organ. According to its onset and pathogenesis, graft rejection is classified into three main types: hyperacute, acute, and chronic.



Hyperacute Rejection


Hyperacute rejection (HAR) occurs within a few minutes to a few hours after the reperfusion of the organ and results in irreversible graft loss. It is triggered by preformed antibodies (donor’s HLA or ABO blood group antigens) directed against antigens presented by the graft. These antibodies activate a series of events that result in diffuse formation of microvascular thrombi, causing ischemic necrosis of the graft [6]. Currently, no treatment is available; however, tissue crossmatching of donor and potential transplant recipients should prevent nearly all HAR.

The panel-reactive antibody (PRA) assay is a screening test that examines the ability of serum from potential transplant recipients to lyse lymphocytes from a panel of HLA-typed donors. A numerical value, expressed as a percentage, indicates the likelihood of a positive crossmatch to the donor population. The finding of a higher PRA identifies patients at high risk for a positive crossmatch and thus for HAR and subsequent graft loss.

Fortunately, pretransplant blood group typing and crossmatching have virtually eliminated the incidence of hyperacute rejection.


Acute Rejection


AR is the most common form of graft rejection. It may develop at any time, but is most frequent during the first several months posttransplant. If it occurs within the first several days posttransplant, the rejection is most likely a combination of amnestic immune response driven by sensitized memory B lymphocytes and activation of the direct allorecognition pathway. This constitutes a rare process termed accelerated acute rejection (AAR) in which the donor antigen exposure often occurred in the distant past. Once the host’s immune systems are challenged by the same donor antigens introduced by the organ transplant, dormant memory lymphocytes reactivate, replicate, and differentiate resulting in graft rejection.

According to the mechanism involved, AR is further divided into cellular (T-cell mediated) rejection, humoral (antibody mediated) rejection, or a combination of both. Unless the host immune system is suppressed pharmacologically, a transplant inevitably leads to acute rejection.

The diagnosis is based on the results of biopsies, special immunologic stains, and laboratory tests (elevated creatinine levels in kidney transplant patients, elevated liver function values in liver transplant recipients, and elevated levels of amylase, and lipase in pancreas transplant hosts).

A variety of immunosuppressive agents are used to prevent AR that will be discussed in the next section.


Chronic Rejection


CR constitutes a slow type of rejection resulting in fibrosis and loss of graft function. The mechanism is not well understood; however, contribution of immune and nonimmune factors is likely. The observation that AR episodes significantly increase the likelihood of CR [7], as well as the correlation observed in renal transplant recipients between poor response to AR treatment and the subsequent development of CR, is evidence of immune factors contributing to this type of rejection. Nonimmune factors include the toxic effects of immunosuppressive agents and cumulative injury from infection (i.e., CMV and polyomavirus) [8, 9]. With advances in immunosuppression, this relatively rare form of rejection is becoming more common.


Immunosuppression Therapy


Balance between the recipient’s immune response, the donor’s allograft, and adequate pharmacologic immunosuppression is necessary for a successful transplant. The initial attempts to establish an effective immunosuppression involved use of total body radiation and 6-mercaptopurine. Results were discouraging, and toxicity was profound. In the early 1960s, the introduction of maintenance immunosuppression through a combination of corticosteroids and azathioprine (a less toxic derivate of 6-mercaptopurine) [10] and the development of cyclosporine, in the 1980s, increased allograft survival [11, 12]. Finally, the introduction of mycophenolate mofetil (MMF) [13] and the calcineurin inhibitor tacrolimus [14] in the 1990s changed the field, enabling a variety of combinations to be used for immunosuppression. Immunosuppressive drugs by groupings are shown in Table 45.1.


Table 45.1
Immunosuppressive drugs by grouping





























































Immunophilin binders

• Calcineurin inhibitors

Cyclosporine

Tacrolimus

• Noninhibitors of calcineurin

Sirolimus

Antimetabolites

• Inhibitors of de novo purine synthesis

Azathioprine

Mycophenolate mofetil

Biologic immunosuppression

• Polyclonal antibodies

ATGAM

Antithymocyte immunoglobulin

• Monoclonal antibodies

Muromonab-CD3 (OKT3)

Basiliximab

Alemtuzumab

Rituximab

Eculizumab

Bortezomib

Fusion proteins

Costimulation-based agents

Belatacept

Other agents

Leflunomide and malononitrilamide (FK778)

Janus Kinase 3 inhibitors

Corticosteroids


Immunosuppressive Strategies


Immunosuppressive agents can be categorized according to their use:



  • Induction—those used for a limited interval at the time of transplant.


  • Maintenance—those used for long term for maintenance of immunosuppression.


  • Antirejection—those used for a short time or in high doses to reverse an acute rejection episode.

Considerably overlap exists among these categories and no single approach applies uniformly. These strategies must take into account the risk of an acute rejection episode, the consequences of an acute rejection episode, and the consequences of a graft loss. The relative importance of each factor varies depending on the organ transplanted.


Pharmacologic Immunosuppression



Immunophilin Binders



Calcineurin Inhibitors


Cyclosporine (CSA) and Tacrolimus (TAC) interfere with the cellular pathway for cytokine production and proliferation. The protein calcineurin has been validated as part of the calcium-dependent signal transduction pathway of interleukin-2 (IL-2) production in T cells. Cyclosporine and Tacrolimus bind to two different intracellular receptors. The resulting receptor complex binds to calcineurin, blocking its phosphatase ability and thereby stopping the production of IL-2 [15].


Cyclosporine


CSA was isolated from a soil sample in Norway and produced by the fungus Tolypocladium inflatum. The introduction of cyclosporine in the early 1980s dramatically altered the field of transplantation by significantly improving outcomes after kidney transplants. CSA binds to its cytoplasmic receptor protein, cyclophilin A (CypA), which subsequently inhibits the activity of calcineurin. T-cell suppression is a result of a decreased expression of several critical T-cell activation genes, the most important being for IL-2 [16]. The first formulation approved by the US Food and Drug Administration (FDA) was Sandimmune® (Novartis, Basel, Switzerland), an oil-based formulation, with poor bioavailability and variable absorption. Newer formulations, Gengraf® (Abbott Laboratories, North Chicago, IL) and Neoral® (Novartis, Basel, Switzerland), are microemulsified making absorption much more reliable. CSA can be given both intravenously or orally to maintain trough levels of 250–350 ng/mL for the first 3 months posttransplant; it can be then tapered to 150–250 ng/mL [17].

CSA is metabolized by the cytochrome P450 3A4 enzyme system that is not only in the liver but also in the cells lining the intestine, so CSA levels can be increased or decreased by changes in gut absorption or in liver metabolism. The extensive side effect profile of CSA has long been a reason for attempts at minimizing drug exposure. Drug interactions and side effects are summarized in Table 45.2. Of most concern is its acute and chronic nephrotoxicity. Acute nephrotoxicity from CSA initially is characterized by vasoconstriction of the afferent arteriole in a dose-dependent, reversible manner. Other side effects include hyperkalemia, hypomagnesemia, hypertension, and neurotoxicity [18]. Cosmetic effects, including hirsutism and gingival hyperplasia, are also observed with the use of CSA.


Table 45.2
Side effects and drug interactions of the main immunosuppressive drugs



















































 
Common side effects

Other medications that increase blood levels

Other medications that decrease blood levels

Other medications that potentiate toxicity

Cyclosporine (CSA)

Hypertension, nephrotoxicity, hirsutism, neurotoxicity, gingival hyperplasia, hypomagnesemia, hyperkalemia

Verapamil, diltiazem, clarithromycin, azithromycin, erythromycin, azole antifungals, protease inhibitors, grapefruit juice

Isoniazid, carbamazepine, phenobarbital, phenytoin, rifampin, St. John’s Wort

Nephrotoxicity: ganciclovir, aminoglycosides, NSAIDs, ACE-Is, and ARBs

Tacrolimus (FK 506)

Hypertension, nephrotoxicity, alopecia, hyperglycemia, neurotoxicity, hypomagnesemia, hyperkalemia

Verapamil, diltiazem, clarithromycin, azithromycin, erythromycin, azole antifungals, protease inhibitors, grapefruit juice

Isoniazid, carbamazepine, phenobarbital, phenytoin, rifampin, St. John’s Wort

Nephrotoxicity: ganciclovir, aminoglycosides, NSAIDs, ACE-Is, and ARBs

Sirolimus

Thrombocytopenia and neutropenia, elevated cholesterol, extremity edema, impaired wound healing

Verapamil, diltiazem, clarithromycin, azithromycin, erythromycin, azole antifungals, protease inhibitors, grapefruit juice

Isoniazid, carbamazepine, phenobarbital, phenytoin, rifampin, St. John’s Wort

__

Mycophenolate mofetil

Leukopenia, thrombocytopenia, GI upset

__

Cholestyramine, antacids

Bone marrow suppression: valganciclovir ganciclovir, TMP/SMX

Corticosteroids

Hyperglycemia, osteoporosis, cataracts, myopathy, weight gain

__

__

__

Azathioprine

Leukopenia, anemia, thrombocytopenia, neoplasia, hepatitis, cholestasis

__

__

Bone marrow suppression: allopurinol, sulfonamides


Tacrolimus


With the success of CSA, researchers studied soil samples from around the world, looking for another compound that might turn out to display immunosuppressive properties. Tacrolimus (TAC), previously known as FK-506, was isolated from a soil sample in Japan in 1984 and produced by the fungus Streptomyces tsukubaensis [19]. Its effect on the lymphocyte is remarkably similar to CSA even though TAC has a completely different chemical structure. TAC acts by binding to FK-binding protein 12(FKBP12) causing inhibition of IL-2 production. The FKP12-TAC complex is 10–100 times more potent than CSA, possibly due to greater affinity for its binding protein [20]. TCA was approved by the FDA in 1994 for the use in liver transplantation. This has been extended to include kidney, heart, small bowel, pancreas, lung, trachea, skin, cornea, bone marrow, and limb transplants, making this drug the backbone of immunosuppressive regimens. TAC can be given intravenously, orally, or sublingually to maintain trough levels of 8–12 ng/mL for the first 3 months posttransplant; then it can be tapered to 6–10 ng/mL.

TAC metabolism is similar to CSA, via the cytochrome P450 34A system. Drug interactions are summarized in Table 45.2. Side effects include alopecia, nephrotoxicity, neurotoxicity, hypertension, hyperkalemia, hypomagnesemia, and new-onset diabetes posttransplant [21].


Noninhibitors of Calcineurin



Sirolimus


Sirolimus (SRL) belongs to a class of compounds known as the mammalian target of rapamycin (mTOR) inhibitors. It is produced by Streptomyces hygroscopicus, a fungus isolated from a soil sample found on Easter Island. SRL, formerly known as rapamycin, was approved in 1999 by the FDA. A derivative of sirolimus, everolimus, was approved by the FDA in 2004. In response to proliferation signals provided by cytokines like IL-2, mTOR, a key regulatory kinase, changes cells from the G1 to S phase in the cell cycle [22]. The mTOR inhibitors bind to FKBP12 (same binding protein as TAC) and this complex causes inhibition of mTOR preventing the cell cycle progression from G1 to S in lymphocytes. The primary pathway for metabolism is the cytochrome P450 34A enzyme system. SRL has been used in a variety of combinations for maintenance of immunosuppression, to help withdraw, or completely avoid, the use of steroids and also as an alternative in calcineurin-sparing protocols. Drug interactions are summarized in Table 45.2. Hypertriglyceridemia is one of the most significant side effects; impaired wound healing, thrombocytopenia, leukopenia, and anemia are also associated with SRL [23, 24].


Antiproliferative Agents


Antiproliferative agents have been part of transplant protocols since the infancy of immunosuppression.


Inhibitors of De Novo Purine Synthesis



Azathioprine


Azathioprine (AZA), a prodrug of 6-mercaptopurine, was developed in the early 1960s and was part of the first successful transplant series. AZA inhibits the purine synthesis in the de novo pathway. Purine inhibition leads to inhibition of the mixed lymphocyte reaction, and to a lesser extent, the antigen antibody reaction [25]. The use of AZA has decreased significantly with the development of new agents such as mycophenolate mofetil (MMF). However, AZA is still preferred in recipients who are considering conceiving a child, because of the teratogenic effects of MMF. The most common side effect is myelosuppression due to suppression of purine synthesis. Most patients can tolerate this effect by reducing the daily dosage, although some need to discontinue the drug entirely. Other significant side effects include hepatotoxicity, pancreatitis, neoplasia, anemia, and pulmonary fibrosis. Severe pancytopenia has been reported when AZA and allopurinol are used together. It is recommended that AZA doses be reduced by 75 % if allopurinol is added to the patient’s drug regimen [25, 26].


Mycophenolate Mofetil


MMF was approved by the FDA in 1995 to prevent acute rejection in kidney recipients. It has been a major addition to the immunosuppressive arsenal and has been incorporated into routine maintenance regimens after many solid organ transplants. MMF is the prodrug of mycophenolic acid (MPA), derived from Penicillium fungi. Mycophenolic acid acts as a noncompetitive inhibitor of inosine monophosphate dehydrogenase, thereby blocking de novo purine synthesis and proliferation in the T and B lymphocytes [13]. MFF is available in capsules; the oral bioavailability approaches 100 %. MPA is glucuronidated in the liver to inactive metabolic mycophenolic acid glucuronide. The most commons side effects of MMF are GI in nature; these include nausea, vomiting, diarrhea, abdominal pain, and gastroesophageal reflux. Neutropenia and thrombocytopenia can also occur with MMF, requiring a dosage reduction. Since MMF is not metabolized by the cytochrome P450 system, interactions only affect its absorption, enterohepatic cycling, or renal excretion [27].


Biologic Immunosuppression


Polyclonal antibodies directed against lymphocytes have been used in transplantation since the 1960s. Monoclonal antibody techniques were discovered later and, in turn, allowed for the development of biologic agents such as OKT3, which target specific subsets of cells. One disadvantage of early murine-based antibody preparations such as OKT3 is the potential for the development of anti-mouse antibodies by the recipient, which may then limit further use of the agent. To address this problem, efforts have focused on the development of humanized versions of monoclonal antibodies (mAbs). These humanized mAbs have a very long half-life, reduced immunogenicity, and the possibility for repeated use [28].


Polyclonal Antibodies


Polyclonal antibodies are produced by immunizing animals, such as horses or rabbits, with human lymphoid tissue, allowing for an immune response, removing the resulting immune sera, and purifying the sera in an effort to remove unwanted antibodies. What remain are antibodies that recognize human lymphocytes. After administration of these antibodies, the transplant recipient’s total lymphocyte count should fall and hence these are known as depleting antibodies.


ATGAM


ATGAM is a purified gamma globulin obtained by immunizing horses with human thymocytes. Infusion into a peripheral vein is often associated with thrombophlebitis; thus, infusion into a central vein is recommended. Side effects, which are more likely related to the release of pyrogenic cytokines, include fever, chills, arthralgia, thrombocytopenia, leukopenia, and serum-like sickness [29]. To avoid the cytokine release syndrome, recipients should be premedicated with methylprednisolone (MP) and diphenhydramine hydrochloride.


Thymoglobulin (ATG-R)


Thymoglobulin is obtained by immunizing rabbits with human thymocytes. Initial kidney transplant studies show ATG-R to be statistically superior to ATGAM in preventing and reversing acute rejection episodes [30]. Administration before reperfusion is advocated to maximize antiadhesion molecules effects. Medications should be given to prevent cytokine release syndrome. Side effects include fever, chills, arthralgias, infections, thrombocytopenia, and leukopenia [29, 31].


Monoclonal Antibodies


Hybridization of murine antibody—secreting B lymphocytes with a nonsecreting myeloma cell line—produces mAbs. Monoclonal antibodies have monovalent affinity, in that they bind to the same epitope.


Muromonab-CD3 (OKT3)


On binding to CD3, OKT3 mediates complement-dependent cell lysis and antibody-dependent cell cytotoxicity leading to rapid clearance of T cells from peripheral circulation [28]. Along with T cell depletion, the overall effect of OKT3 is likely to be due to interrupted T cell receptor binding and internalization, disrupted trafficking, and cytokine-mediated regulatory changes. If OKT3 is effective, the percentage of CD3-positive cells in the circulation should fall to, and stay below, 5 %. Side effects may occur when cytokines (e.g., TNF, IL-2, and interferon) are released by T cells into the circulation. Premedication with MP and diphenhydramine hydrochloride is important to minimize these side effects. The most serious side effect with OKT3 is a rapidly developing, noncardiogenic pulmonary edema that can be life threatening [32]. Other side effects include neurotoxicity, nephrotoxicity, and allograft thrombosis.


Anti-Interleukin-2 Monoclonal Antibodies (Basiliximab)


Basiliximab is an anti-CD25 monoclonal antibody. The alpha subunit of the IL-2 receptor, also known as Tac or CD25, is found exclusively on activated T cells. Binding of these agents to the IL-2 receptor results in blockade of IL-2-mediated responses. Because major portions of these agents are of human origin, they tend to have much longer half-lives than does OKT3. No lymphocyte depletion occurs with basiliximab; it is not designed to treat acute rejection. Its selectivity in blocking IL-2-mediated responses makes it a powerful induction agent without the added risks of infections, malignancies, or other major side effects [33, 34].


Alemtuzumab


Alemtuzumab is a CD52-specific humanized monoclonal antibody, initially used to treat chronic lymphocytic leukemia. It rapidly depletes CD52 expressing lymphocytes centrally and peripherally resulting from bulk T cell depletion with lesser depletion of B cells and monocytes. Alemtuzumab facilitates reduced maintenance immunosuppression requirements without an increase in infections or malignant complications in solid organ transplantations as compared to historical controls. Alemtuzumab causes a significant cytokine release reaction and often requires premedication [35, 36].


Rituximab


Rituximab is a chimeric monoclonal antibody specific for CD20, a cell surface glycoprotein involved in B cell activation and maturation [37]. It is FDA approved for treating lymphoma; however, its use has grown to include the treatment of antibody-mediated rejection and use in desensitization protocols with plasmapheresis and/or intravenous immunoglobulin [38]. The most important indication of Rituximab in solid organ transplantation is as a primary treatment of PTLD.


Eculizumab


Eculizumab is a humanized monoclonal antibody that is a first-in-class terminal complement inhibitor, with high affinity for C5, and the first therapy approved for the treatment of paroxysmal nocturnal hemoglobinuria (PNH). It blocks the activation of terminal complement. Adverse effects include an increased risk of infections, especially from encapsulated bacteria.


Bortezomib


Bortezomib is a proteasome inhibitor, FDA approved for treating myeloma. It has been shown to cause apoptosis of normal plasma cells, thereby decreasing alloantibody production in sensitized patients. Several case reports and series have described the use of Bortezomib for the treatment of antibody-mediated rejection and in desensitization protocols [3840].


Fusion Proteins


Fusion proteins are made by the fusion of a single receptor targeting a ligand of interest with a secondary molecule, which is typically the Fc portion of an IgG molecule.


Costimulation-Based Agents


Costimulatory molecules alter the threshold for activation of naive T lymphocytes without having a primary activating or inhibitory function. The two costimulatory receptors on T cells are CD28 and CD152; these serve reciprocal roles: CD28 facilitates a T cell response, whereas CD152 reduces it.


Belatacept


Belatacept (also known as LEA29Y) is a second-generation costimulation-blockade agent that has two amino acid substitutions. It is a high-avidity molecule with slower dissociation rates. Belatacept is a promising, nonnephrotoxic option in kidney transplant recipients and is being developed with the aim of providing calcineurin inhibitors avoidance [41, 42].


Other Immunosuppressive Agents



Leflunomide and Malononitrilamide (FK778)


Leflunomide and its analogues have strong antiproliferative effects on T lymphocytes and B lymphocytes. This group of medications also acts through inhibition of tyrosine kinase. FK778 and Leflunomide possess antiviral effects and have been used successfully to treat cytomegalovirus and BK virus nephropathy in renal transplant patients [43, 44].


Janus Kinase 3 Inhibitors


Janus kinase 3 (JAK3) is essential for the signal transduction from the cytokine receptors of several cytokines to the nucleus. JAK3 is expressed on immune cells only, making it an important target for developing new immunosuppressants. CP-690559 is the most potent and selective JAK3 inhibitor. Its effects include reduction in natural killer cell and T cell numbers, with unchanged numbers of CD8+ effector memory T cells [45].


Corticosteroids


Steroids have been a part of transplantation since its inception. It soon became clear, however, that the toxicities of steroids could overshadow their benefits. Despite numerous attempts to limit or discontinue their use, they remain an integral component of most immunosuppressive protocols. Steroids inhibit T cell proliferation, T cell-dependent immunity, and the expression of various cytokines, especially IL-2, IL-6, and interferon-γ, and tumor necrosis factor-α (TNF-α). They also suppress antibody formation and the delayed hypersensitivity response found in allograft rejection. Steroids use is associated with a series of problems, including acne, increased appetite, mood changes, diabetes, hypertension, and impaired wound healing [46]. Many transplant centers are switching to steroid-withdrawal/steroid-free protocols for many of their recipients [47, 48].

Only gold members can continue reading. Log In or Register to continue

Oct 28, 2016 | Posted by in CRITICAL CARE | Comments Off on Advances in Immunosuppressive Therapy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access