You are on page 1of 15

The new england journal of medicine

review article

drug therapy

Immunosuppressive Drugs for Kidney


Transplantation
Philip F. Halloran, M.D., Ph.D.

t he central issue in organ transplantation remains suppres-


sion of allograft rejection. Thus, development of immunosuppressive drugs is
the key to successful allograft function. Immunosuppressive agents are used
for induction (intense immunosuppression in the initial days after transplantation),
maintenance, and reversal of established rejection. This review focuses on agents that
From the Division of Nephrology and Trans-
plantation Immunology, University of Alber-
ta, Edmonton, Canada. Address reprint re-
quests to Dr. Halloran at 250 Heritage
Medical Research Centre, Edmonton, AB
T6G 2S2, Canada, or at phil.halloran@
ualberta.ca.
are either approved or in phase 2 or phase 3 trials in kidney transplantation, but many
issues covered here are applicable to all organ transplantation. I begin with a model of N Engl J Med 2004;351:2715-29.
Copyright © 2004 Massachusetts Medical Society.
the alloimmune response to illustrate how these medications act.

three-signal model of alloimmune responses

Alloimmune responses involve both naive and memory lymphocytes,1 including lym-
phocytes previously stimulated by viral antigens cross-reacting with HLA antigens.2 In
the graft and the surrounding tissues, dendritic cells of donor and host origin become
activated and move to T-cell areas of secondary lymphoid organs. There, antigen-bear-
ing dendritic cells engage alloantigen-reactive naive T cells and central memory T cells
that recirculate between lymphoid compartments but cannot enter peripheral tissues3
(Fig. 1). Naive T cells are optimally triggered by dendritic cells in secondary lymphoid
organs,6,7 but antigen-experienced cells may be activated by other cell types, such as
graft endothelium.8
An antigen on the surface of dendritic cells that triggers T cells with cognate T-cell
receptors constitutes “signal 1,” transduced through the CD3 complex. Dendritic cells
provide costimulation, or “signal 2,” delivered when CD80 and CD86 on the surface of
dendritic cells engage CD28 on T cells. Signals 1 and 2 activate three signal transduc-
tion pathways: the calcium–calcineurin pathway, the RAS–mitogen-activated protein
(MAP) kinase pathway, and the nuclear factor-kB pathway.9 These pathways activate
transcription factors that trigger the expression of many new molecules, including in-
terleukin-2, CD154, and CD25. Interleukin-2 and other cytokines (e.g., interleukin-15)
activate the “target of rapamycin” pathway to provide “signal 3,” the trigger for cell pro-
liferation. Lymphocyte proliferation also requires nucleotide synthesis. Proliferation
and differentiation lead to a large number of effector T cells. B cells are activated when
antigen engages their antigen receptors, usually in lymphoid follicles or in extrafollicu-
lar sites, such as red pulp of spleen,10 or possibly in the transplant,11 producing alloan-
tibody against donor HLA antigens. Thus, within days the immune response generates
the agents of allograft rejection, effector T cells and alloantibody.

effectors and lesions of rejection


Effector T cells that emerge from lymphoid organs infiltrate the graft and orchestrate
an inflammatory response. In T-cell–mediated rejection, the graft is infiltrated by effec-

n engl j med 351;26 www.nejm.org december 23, 2004 2715

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

tor T cells, activated macrophages, B cells, and plas-


ma cells and displays interferon-g effects, increased Figure 1 (facing page). Steps in T-Cell–Mediated Rejection.
chemokine expression, altered capillary permeabili- Antigen-presenting cells of host or donor origin migrate
to T-cell areas of secondary lymphoid organs. These
ty and extracellular matrix, and deterioration of T cells ordinarily circulate between lymphoid tissues,
parenchymal function. The diagnostic lesions of regulated by chemokine and sphingosine-1-phosphate
T-cell–mediated rejection reflect mononuclear cells (S-1-P) receptors.4 APCs present donor antigen to naive
invading the kidney tubules (tubulitis) and the inti- and central memory T cells. Some presentation of antigen
ma of small arteries (arteritis). Macrophages that by donor cells in the graft cannot be excluded (e.g., endo-
thelial cells that activate antigen-experienced T cells).
are activated by T cells participate through delayed- T cells are activated and undergo clonal expansion and
type hypersensitivity,12 but the injury remains anti- differentiation to express effector functions. Antigen trig-
gen-specific.13 Injury is not simply lysis of target gers T-cell receptors (TCRs) (signal 1) and synapse for-
cells, since typical lesions develop in mice lacking mation.5 CD80 (B7-1) and CD86 (B7-2) on the APC engage
cytotoxic T-cell lytic molecules,14 but may involve CD28 on the T cell to provide signal 2. These signals acti-
vate three signal-transduction pathways — the calcium–
parenchymal transdifferentiation into mesenchy- calcineurin pathway, the mitogen-activated protein (MAP)
mal cells15 and cell senescence.16 kinase pathway, and the protein kinase C–nuclear factor-
Alloantibody against donor antigens that is pro- kB (NF-kB) pathway — which activate transcription fac-
duced systemically or locally in the graft targets tors nuclear factor of activated T cells (NFAT), activating
capillary endothelium.17 Antibody-mediated rejec- protein 1 (AP-1), and NF-kB, respectively. The result is
expression of CD154 (which further activates APCs), in-
tion is diagnosed by clinical,18 immunologic,19 and terleukin-2 receptor a chain (CD25), and interleukin-2.
histologic criteria, including a demonstration of Receptors for a number of cytokines (interleukin-2, 4,
complement factor C4d in capillaries.20 7, 15, and 21) share the common g chain, which binds
Janus kinase 3 (JAK3). Interleukin-2 and interleukin-15
host–graft adaptation deliver growth signals (signal 3) through the phosphoinosit-
ide-3-kinase (PI-3K) pathway and the molecular-target-
The term “host–graft adaptation” describes the de- of-rapamycin (mTOR) pathway, which initiates the cell
crease in both donor-specific responsiveness and cycle. Lymphocytes require synthesis of purine and pyri-
the risk of rejection in the months after a successful midine nucleotides for replication, regulated by inosine
transplantation that is maintained with immuno- monophosphate dehydrogenase (IMPDH) and dihydro-
suppression.21 Changes in the organ — a loss of orotate dehydrogenase (DHODH), respectively. Antigen-
experienced T cells home to and infiltrate the graft and
donor dendritic cells and a resolution of injury — engage the parenchyma to create typical rejection lesions
contribute to the adaptation. Regulatory T cells may such as tubulitis and, in more advanced rejection, endo-
also be able to control alloimmune responses, by thelial arteritis. However, if the rejection does not destroy
analogy with their ability to suppress autoimmuni- the graft, adaptation occurs and is stabilized by immuno-
ty,22 although this hypothesis is unproven. The cru- suppressive drugs. The photomicrographs of tubulitis
and endothelial arteritis are taken from a mouse model
cial element is that host T cells become less respon- in which these lesions are T-cell–dependent but indepen-
sive to donor antigens when antigen persists and dent of perforin, granzymes, and antibody. IKK denotes
immunosuppression is maintained. This may be a inhibitor of nuclear factor-kB kinase , CDK cyclin-depen-
general characteristic of T-cell responses in vivo, in dent kinase, and MHC major histocompatibility complex.
which antigen persistence with inadequate costim-
ulation triggers adaptations that limit T-cell re-
sponsiveness.23 The resulting partial T-cell anergy tions that calcineurin inhibitors prevent adapta-
(known as “adaptive tolerance” or “in vivo anergy”) tions in clinical transplantation. However, the rele-
is characterized by decreased tyrosine kinase acti- vance of these models to clinical adaptation, which
vation and calcium mobilization (signal 1) and de- occurs despite treatment with calcineurin inhibi-
creased response to interleukin-2 (signal 3). Adap- tors, is doubtful.
tation in clinical transplantation resembles in vivo
anergy — for example, both can occur in the pres- immunosuppressive drugs
ence of calcineurin inhibitors. The role of mainte-
nance immunosuppression may be to stabilize ad- Immunosuppression can be achieved by deplet-
aptation by limiting excitation of the immune system ing lymphocytes, diverting lymphocyte traffic, or
and thus antigen presentation. In some experi- blocking lymphocyte response pathways (Fig. 2).
mental models, favorable adaptations are blocked Immunosuppressive drugs have three effects: the
when calcineurin is inhibited,24 leading to sugges- therapeutic effect (suppressing rejection), unde-

2716 n engl j med 351;26 www.nejm.org december 23 , 2004

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

n engl j med 351;26 www.nejm.org december 23, 2004 2717

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

Figure 2. Individual Immunosuppressive Drugs and Sites of Action in the Three-Signal Model.
Anti-CD154 antibody has been withdrawn from clinical trials but remains of interest. FTY720 engagement of sphingosine-1-phosphate (S-1-P)
receptors triggers and internalizes the receptors and alters lymphocyte recirculation, causing lymphopenia. Antagonists of chemokine recep-
tors (not shown) are also being developed in preclinical models. MPA denotes mycophenolic acid.

sired consequences of immunodeficiency (infec- ten related to the mechanism that is used, because
tion or cancer), and nonimmune toxicity to other tis- each agent or class of drugs targets molecules with
sues. Immunodeficiency leads to characteristic physiologic roles in nonimmune tissues. For ex-
infections and cancers, such as post-transplanta- ample, nephrotoxicity of calcineurin inhibitors may
tion lymphoproliferative disease,25 which are relat- reflect a role of calcineurin within the renal vas-
ed more to the intensity of immunosuppression culature.
than to the specific agent used.
New immunosuppressive protocols underscored classification of immunosuppressive
this point by evoking a new infectious complica- drugs
tion, BK-related polyomavirus nephropathy.26 This Immunosuppressive drugs include small-molecule
syndrome of tubular injury by a virus that is usually drugs, depleting and nondepleting protein drugs
innocuous emerged only with the recent introduc- (polyclonal and monoclonal antibodies), fusion pro-
tion of powerful drug combinations and now con- teins, intravenous immune globulin, and glucocor-
tributes to renal injury and graft loss. Fortunately, ticoids (Table 1). Because of space limitations, in-
the newer immunosuppressive agents have result- travenous immune globulin and glucocorticoids
ed in a lower incidence of both infection and cancer cannot be discussed in detail. In brief, intravenous
than might have been expected, perhaps because immune globulin has multiple effects27 and is an
preventing rejection reduces the need for powerful important component of approaches to suppress
agents to reverse it. alloantibody responses. Glucocorticoids act as ago-
Nonimmune toxicity is agent-specific and is of- nists of glucocorticoid receptors, but at higher dos-

2718 n engl j med 351;26 www.nejm.org december 23 , 2004

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

es they have receptor-independent effects. Recep-


Table 1. Classification of Immunosuppressive Therapies
tor-mediated effects are mainly transcriptional
Used in Organ Transplantation or in Phase 2–3 Trials.*
through DNA-binding and protein–protein inter-
actions of the steroid-receptor complex, targeting Glucocorticoids
transcription factors such as activator protein 1 and Small-molecule drugs
Immunophilin-binding drugs
nuclear factor-kB.28 Calcineurin inhibitors
Most small-molecule immunosuppressive agents Cyclophilin-binding drugs: cyclosporine,
are derived from microbial products and target pro- ISA(TX)247†
FKBP12-binding drugs: tacrolimus, modified-
teins that have been highly conserved in evolution. release tacrolimus‡
Small-molecule immunosuppressive drugs at clin- Target-of-rapamycin inhibitors: sirolimus,
ically tolerated concentrations probably do not sat- everolimus
Inhibitors of nucleotide synthesis
urate their targets. For example, cyclosporine acts Purine synthesis (IMPDH) inhibitors
by inhibiting calcineurin but only partially inhibits Mycophenolate mofetil
calcineurin as used clinically.29 Without target sat- Enteric-coated mycophenolic acid
Mizoribine§
uration, the drug’s effects are proportional to the Pyrimidine synthesis (DHODH) inhibitors
concentration of the drug, which makes dosing and Leflunomide¶
monitoring critical. FK778†
Antimetabolites: azathioprine
Depleting protein immunosuppressive agents Sphingosine-1-phosphate–receptor antagonists:
are antibodies that destroy T cells, B cells, or both. FTY720‡
T-cell depletion is often accompanied by the re- Protein drugs
lease of cytokines, which produces severe systemic Depleting antibodies (against T cells, B cells, or both)
Polyclonal antibody: horse or rabbit antithymo-
symptoms, especially after the first dose. The use cyte globulin
of depleting antibodies reduces early rejection but Mouse monoclonal anti-CD3 antibody (muromo-
increases the risks of infection and post-trans- nab-CD3)
Humanized monoclonal anti-CD52 antibody
plantation lymphoproliferative disease and can be (alemtuzumab)¶
followed by late rejection as the immune system B-cell–depleting monoclonal anti-CD20 antibody
recovers. Recovery from immune depletion takes (rituximab)¶
Nondepleting antibodies and fusion proteins
months to years and may never be complete in older Humanized or chimeric monoclonal anti-CD25
adults. The depletion of antibody-producing cells antibody (daclizumab, basiliximab)
is better tolerated than T-cell depletion, because it Fusion protein with natural binding properties:
CTLA-4–Ig (LEA29Y†)
is not usually accompanied by cytokine release and Intravenous immune globulin
immunoglobulin levels are usually maintained.
However, depletion of antibody-producing cells is * FKBP12 denotes FK506-binding protein 12, IMPDH ino-
incomplete because many plasma cells are resis- sine monophosphate dehydrogenase, DHODH dihydro-
orotate dehydrogenase, and CTLA-4 cytotoxic T-lympho-
tant to the available antibodies that target B cells, cyte–associated antigen 4.
such as anti-CD20 antibody. † This treatment is being used in phase 2 trials in renal
Nondepleting protein drugs are monoclonal transplantation.
‡ This treatment is being used in phase 3 trials in renal
antibodies or fusion proteins that reduce respon- transplantation.
siveness without compromising lymphocyte popu- § Mizoribine is being used as an immunosuppressive drug
lations. They typically target a semiredundant mech- in Japan.
¶ This drug is being evaluated for off-label use as an immu-
anism such as CD25, which explains their limited nosuppressive agent.
efficacy but the absence of immunodeficiency com-
plications. These drugs have low nonimmune tox-
icity because they target proteins that are expressed
only in immune cells and trigger little release of cy- Gertrude Elion and George Hitchings, were ac-
tokines. knowledged by a share of the 1988 Nobel Prize.
Azathioprine is thought to act by releasing 6-mer-
small-molecule drugs captopurine, which interferes with DNA synthesis.
Azathioprine, which is derived from 6-mercapto- Other possible mechanisms include converting co-
purine, was the first immunosuppressive agent stimulation into an apoptotic signal.41 After cyclo-
to achieve widespread use in organ transplanta- sporine was introduced, azathioprine became a sec-
tion30 (Table 2). The developers of azathioprine, ond-line drug.

n engl j med 351;26 www.nejm.org december 23, 2004 2719

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

Table 2. Characteristics of Small-Molecule Immunosuppressive Drugs Used in Organ Transplantation or in Phase 2–3 Trials.*

Drug Description Mechanism Nonimmune Toxicity and Comments

Cyclosporine 11-amino-acid cyclic Binds to cyclophilin; complex inhibits Nephrotoxicity, hemolytic–uremic syndrome, hyperten-
peptide from Tolypo- calcineurin phosphatase and T-cell sion, neurotoxicity, gum hyperplasia, skin changes,
cladium inflatum31 activation hirsutism, post-transplantation diabetes mellitus,
hyperlipidemia; trough monitoring or checking lev-
els two hours after administration required
Tacrolimus Macrolide antibiotic Binds to FKBP12; complex inhibits Effects similar to those of cyclosporine but with a lower in-
(FK506) from Streptomyces calcineurin phosphatase and T-cell cidence of hypertension, hyperlipidemia, skin chang-
tsukubaensis32,33 activation es, hirsutism, and gum hyperplasia and a higher inci-
dence of post-transplantation diabetes mellitus and
neurotoxicity; trough monitoring required
Sirolimus Triene macrolide antibi- Binds to FKBP12; complex inhibits Hyperlipidemia, increased toxicity of calcineurin inhib-
(rapamycin) otic from S. hygro- target of rapamycin and interleu- itors, thrombocytopenia, delayed wound healing,
scopicus from Easter kin-2–driven T-cell proliferation delayed graft function, mouth ulcers, pneumonitis,
Island (Rapa Nui)34 interstitial lung disease; lipid monitoring required;
recipients whose risk of rejection is low to moderate
can stop cyclosporine treatment two to four months
after transplantation
Everolimus Derivative of sirolimus
Mycophenolate Mycophenolic acid Inhibits synthesis of guanosine mon- Gastrointestinal symptoms (mainly diarrhea), neutro-
mofetil and from penicillium ophosphate nucleotides; blocks penia, mild anemia; blood-level monitoring not re-
enteric-coated molds35-37 purine synthesis, preventing pro- quired but may improve efficacy; absorption reduced
mycophenolate liferation of T and B cells by cyclosporine
FK778 and Modification of A77 Inhibits pyrimidine synthesis, blocking Anemia; other effects not known; in phase 2 trials
malononi- 1726 (active deriva- proliferation of T and B cells
trilamide tive of leflunomide)
Azathioprine Prodrug that releases Converts 6-mercaptopurine to tissue Leukopenia, bone marrow depression, macrocytosis,
6-mercaptopurine inhibitor of metalloproteinase, liver toxicity (uncommon); blood-count monitoring
which is converted to thioguanine required
nucleotides that interfere with DNA
synthesis; thioguanine derivatives
may inhibit purine synthesis
FTY720 Sphingosine-like deriva- Works as an antagonist for sphingo- Reversible first-dose bradycardia, potentiated by general
tive of myriocin sine-1-phosphate receptors on anesthetics and beta-blockers; nausea, vomiting,
from ascomycete lymphocytes, enhancing homing diarrhea, increased liver-enzyme levels
fungus38 to lymphoid tissues and prevent-
ing egress, causing lymphopenia
CP-690,55039; Synthetic molecule Binds cytoplasmic tyrosine kinase Anemia caused by potential effects on JAK2
and Tyrphostin JAK3, inhibiting cytokine-induced
AG 49040 signaling

* Data about drugs come from the manufacturer’s inserts for health care professionals unless otherwise indicated.

Calcineurin Inhibitors the hemolytic–uremic syndrome and post-trans-


Cyclosporine, a cornerstone of immunosuppres- plantation diabetes mellitus. Recent developments
sion in transplantation for two decades, is in effect include monitoring of the peak cyclosporine levels
a prodrug that engages cyclophilin, an intracellular two hours after administration to better reflect ex-
protein of the immunophilin family, forming a com- posure to the drug.43,44A chemically modified cy-
plex that then engages calcineurin.42 The adverse closporine, ISA(TX)247, is under development.45
effects of cyclosporine, which are related to the con- Tacrolimus engages another immunophilin,
centration of the drug, include nephrotoxicity, hy- FK506-binding protein 12 (FKBP12), to create a
pertension, hyperlipidemia, gingival hyperplasia, complex that inhibits calcineurin with greater mo-
hirsutism, and tremor. Cyclosporine can also induce lar potency than does cyclosporine. Initial trials in-

2720 n engl j med 351;26 www.nejm.org december 23 , 2004

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

dicated that there was less rejection with tacrolimus tipneumocystis activity.59 Enteric-coated mycophe-
than with cyclosporine,46,47 but recent analyses sug- nolic acid has been introduced as an alternative to
gest that in the current dosing strategies, the effi- mycophenolate mofetil.60
cacy of cyclosporine is similar to that of tacroli-
mus.48,49 Tacrolimus resembles cyclosporine in that Target-of-Rapamycin Inhibitors
it can result in nephrotoxicity and the hemolytic– Sirolimus61 and everolimus engage FKBP12 to cre-
uremic syndrome, but it is less likely to cause hy- ate complexes that engage and inhibit the target of
perlipidemia, hypertension, and cosmetic problems rapamycin but cannot inhibit calcineurin (Fig. 2).
and more likely to induce post-transplantation dia- Inhibition of the target of rapamycin blocks sig-
betes. Tacrolimus has been suspected of inducing nal 3 by preventing cytokine receptors from activat-
more BK-related polyomavirus nephropathy than ing the cell cycle. The principal nonimmune toxic
has cyclosporine in patients who have undergone effects of sirolimus and everolimus include hyper-
kidney transplantation, especially when used with lipidemia, thrombocytopenia, and impaired wound
mycophenolate mofetil, but renal function may be healing. Other reported effects include delayed re-
better with tacrolimus.49 New developments in- covery from acute tubular necrosis in kidney trans-
clude a preparation of modified-release tacroli- plants, reduced testosterone concentrations,62 ag-
mus to permit once-daily dosing. gravation of proteinuria, mouth ulcers, skin lesions,
The use of tacrolimus has increased steadily, and pneumonitis. However, sirolimus and everoli-
and the drug is now the dominant calcineurin in- mus may reduce cytomegalovirus disease.63 Siroli-
hibitor, but most transplantation programs exploit mus and everolimus were developed for use with
the strengths of both tacrolimus and cyclosporine, cyclosporine,64,65 but the combination increased
depending on the risks in individual patients. Hy- nephrotoxicity, the hemolytic–uremic syndrome,
pertension, hyperlipidemia, and the risk of rejec- and hypertension. Sirolimus has been combined
tion argue for tacrolimus, whereas a high risk of with tacrolimus (e.g., the Edmonton protocol for
diabetes (e.g., older age or obesity) argues for cy- pancreatic islet transplantation) to avoid the toxic-
closporine. ity of sirolimus–cyclosporine combinations.66,67
However, a controlled trial in renal transplantation
Inosine Monophosphate Dehydrogenase Inhibitors showed that sirolimus plus tacrolimus produced
The use of inhibitors of purine synthesis for immu- more renal dysfunction and hypertension than did
nosuppression was based on the observation that mycophenolate mofetil plus tacrolimus,68 which in-
inborn errors in this pathway produce immunode- dicates that sirolimus potentiates tacrolimus neph-
ficiency without damaging other organs, in con- rotoxicity. Practitioners can reduce the toxicity of
trast to errors in the purine salvage pathway.50,51 the combination of a target-of-rapamycin inhibitor
Mycophenolic acid inhibits inosine monophosphate and a calcineurin inhibitor by withdrawing one of
dehydrogenase, a key enzyme in purine synthesis. the drugs. For example, withdrawing cyclosporine
Mycophenolate mofetil is a prodrug that releases in patients in stable condition who are receiving
mycophenolic acid, and in large-scale trials with the sirolimus–cyclosporine combination reduces
cyclosporine, it was superior to azathioprine in pre- renal dysfunction and hypertension, with a small
venting rejection of kidney transplants.52-55 Pro- increase in rejection episodes,69 which suggests a
tocols using mycophenolate mofetil and calcineurin strategy for avoiding the toxic effects of calcineurin
inhibitors improved patient survival and graft sur- inhibitors (Table 3).
vival and reduced early and late allograft rejec- Sirolimus and everolimus may have antineo-
tion.56,57 Mycophenolate mofetil has also been plastic and arterial protective effects. Since these
evaluated in heart transplantation.58 The drug has agents slow the growth of established experimen-
largely replaced azathioprine and is widely used be- tal tumors,70 they have potential applications in
cause it is effective in combination with many oth- oncology. The possibility that sirolimus and everoli-
er agents, simple to use without monitoring, and mus can protect arteries is suggested by two ob-
free from organ toxicity and cardiovascular risk. Its servations: target-of-rapamycin inhibitors that are
principal nonimmune toxic effects are gastrointes- incorporated into coronary stents inhibit resteno-
tinal (mainly diarrhea) and hematologic (anemia, sis,71 and target-of-rapamycin inhibitors plus cal-
leukopenia). Mycophenolate mofetil may increase cineurin inhibitors reduce the incidence of graft
cytomegalovirus disease but in vitro manifests an- coronary artery disease associated with heart trans-

n engl j med 351;26 www.nejm.org december 23, 2004 2721

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

Table 3. Characteristics of Protein Immunosuppressive Drugs Used in Organ Transplantation or in Phase 2–3 Trials.

Drug Description Mechanism Toxicity and Comments*

Polyclonal anti- Polyclonal IgG from horses or rabbits Blocks T-cell membrane proteins (CD2, The cytokine-release syndrome (fever,
thymocyte immunized with human thymo- CD3, CD45, and so forth), causing al- chills, hypotension), thrombocyto-
globulin cytes; absorbed to reduce unwant- tered function, lysis, and prolonged penia, leukopenia, serum sickness
ed antibodies T-cell depletion
Muromonab- Murine monoclonal antibody against Binds to CD3 associated with T-cell recep- Severe cytokine-release syndrome,
CD3 CD3 component of T-cell–recep- tor, leading to initial activation and pulmonary edema, acute renal fail-
tor signal-transduction complex cytokine release, followed by blockade ure, gastrointestinal disturbances,
of function, lysis, and T-cell depletion changes in central nervous system
Alemtuzumab Humanized monoclonal antibody Binds to CD52 on all B and T cells, most Mild cytokine-release syndrome, neu-
against CD52, a 25-to-29-kD monocytes, macrophages, and natu- tropenia, anemia, idiosyncratic pan-
membrane protein ral killer cells, causing cell lysis and cytopenia, autoimmune thrombo-
prolonged depletion cytopenia, thyroid disease
Rituximab Chimeric monoclonal antibody Binds to CD20 on B cells and mediates Infusion reactions, hypersensitivity
against membrane-spanning B-cell lysis reactions (uncommon)
four-domain protein CD20
Basiliximab Chimeric monoclonal antibody Binds to and blocks the interleukin-2– Hypersensitivity reactions (uncommon);
against CD25 (interleukin-2– receptor a chain (CD25 antigen) on two doses required; no monitoring
receptor a chain) activated T cells, depleting them and required
inhibiting interleukin-2–induced T-cell
activation
Daclizumab Humanized monoclonal antibody Has similar action to that of basiliximab Hypersensitivity reactions (uncommon);
against CD25 (interleukin-2– five doses recommended but two
receptor a chain) may suffice; no monitoring required
LEA29Y Protein combining B7-binding portion Binds to B7 on T cells, preventing CD28 Effects unknown; in phase 2 trials
of CTLA-4 with IgG Fc region signaling and signal 2

* The toxic effects of alemtuzumab, rituximab, and LEA29Y in organ-transplant recipients must be established in phase 3 trials. The toxic effects
of alemtuzumab are primarily those reported in nontransplantation trials.

plantation.63 But alternative explanations exist for FTY720


both observations. Target-of-rapamycin inhibitors FTY720 is derived from myriocin, a fungus-derived
may suppress restenosis of mechanically dilated ar- sphingosine analogue. After phosphorylation,
teries by suppressing wound healing72 rather than FTY720 engages lymphocyte sphingosine-1-phos-
by atherogenesis and may prevent graft coronary phate receptors and profoundly alters lymphocyte
artery disease simply by preventing rejection. Po- traffic, acting as a functional sphingosine-1-phos-
tential arterial protective effects of sirolimus and phate antagonist.75 FTY720 drives T cells into lym-
everolimus must be weighed against the effects of phoid tissues and prevents them from leaving and
the hyperlipidemia these drugs induce.73 homing to the graft. Despite low overall toxicity,
FTY720 induces reversible bradycardia during the
Dihydroorotate Dehydrogenase Inhibitors first doses,76 arousing concern about the potential
Dihydroorotate dehydrogenase is a key enzyme in for cardiac arrest when combined with the influ-
pyrimidine synthesis. Leflunomide is a dihydrooro- ences of other agents (e.g., general anesthetics or
tate dehydrogenase inhibitor that is approved for beta-blockers). FTY720 in combination with cyclo-
rheumatoid arthritis but is not widely used in trans- sporine has completed phase 2 trials77 and entered
plantation.74 Its active metabolite, A77 1726, was phase 3 trials in renal transplantation.
modified to create FK778, which is in phase 2 trials
in kidney transplantation. FK778 may have activity depleting antibodies
against BK-related polyomavirus and have a lower Polyclonal antithymocyte globulin is produced by
incidence of gastrointestinal effects than does my- immunizing horses or rabbits with human lym-
cophenolate mofetil, but its nonimmune toxic ef- phoid cells, harvesting the IgG, and absorbing out
fects such as anemia must be evaluated. toxic antibodies (e.g., those against platelets and

2722 n engl j med 351;26 www.nejm.org december 23 , 2004

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

erythrocytes) (Table 3). As an induction agent, poly- trolled trials are needed to establish dosing, safety,
clonal antithymocyte globulin is usually used for and efficacy.
3 to 10 days to produce “profound and durable” Rituximab (anti-CD20 monoclonal antibody)
lymphopenia that lasts beyond one year.78 In addi- eliminates most B cells and is approved for treat-
tion to immunodeficiency complications, toxic ef- ing refractory non-Hodgkin’s B-cell lymphomas,
fects of polyclonal antithymocyte globulin include including some post-transplantation lymphopro-
thrombocytopenia, the cytokine-release syndrome, liferative disease in organ-transplant recipients.
and occasional serum sickness or allergic reactions. Rituximab is used off-label in combination with
Rabbit preparations of polyclonal antithymocyte maintenance immunosuppressive drugs, plasma-
globulin (such as Thymoglobulin and ATG-Frese- pheresis, and intravenous immune globulin to sup-
nius) are favored over horse polyclonal antithy- press deleterious alloantibody responses in trans-
mocyte globulin because of greater potency. plant recipients. Although plasma cells are usually
Muromonab-CD3, a mouse monoclonal anti- CD20-negative, many are short-lived and require
body against CD3, has been used for 20 years to treat replacement from CD20-positive precursors. Thus,
rejection79 and for induction.80 Muromonab-CD3 depletion of CD20-positive cells does reduce some
binds to T-cell-receptor–associated CD3 complex antibody responses. CD20-positive B cells can act
and triggers a massive cytokine-release syndrome as secondary antigen-presenting cells, which raises
before both depleting and functionally altering the possibility that rituximab can ameliorate T-cell
T cells. Humans can make neutralizing antibodies responses. Off-label applications for rituximab in-
against muromonab-CD3 that terminate its effect clude treatment of antibody-mediated rejection and
and limit its reuse. Prolonged courses of muromo- possibly severe T-cell–mediated rejection88 and sup-
nab-CD3 increase the risk of post-transplantation pression of preformed alloantibody before trans-
lymphoproliferative disease.81 The use of muro- plantation. Again, controlled trials are needed.
monab-CD3 declined when newer small-molecule
immunosuppressive drugs reduced rejection epi- nondepleting antibodies and fusion
sodes. A trial of a humanized anti-CD3 mono- proteins
clonal antibody in kidney transplantation82 was Daclizumab and Basiliximab
stopped. (A nonactivating humanized anti-CD3 The anti-CD25 monoclonal antibodies daclizumab
monoclonal antibody is being developed to sup- and basiliximab are widely used in transplantation
press beta-cell injury in patients with autoimmune for induction in patients who have a low-to-moder-
diabetes mellitus of recent onset83 but is not current- ate risk of rejection. Because expression of CD25
ly used for transplantation.) (interleukin-2 receptor a chain) requires T-cell acti-
Alemtuzumab, a humanized monoclonal anti- vation, anti-CD25 antibody causes little depletion
body against CD52, massively depletes lymphocyte of T cells. Anti-CD25 antibody is moderately effec-
populations. It is approved for treating refractory tive since it reduces rejection by about one third
B-cell chronic lymphocytic leukemia but is not ap- when used with calcineurin inhibitors and has min-
proved for immunosuppression in transplantation. imal toxic effects.89-92
A small study in renal transplantation that conclud-
ed that alemtuzumab induced “prope tolerance” LEA29Y
(meaning near-tolerance)84 was not confirmed in LEA29Y is a second-generation cytotoxic-T-lympho-
later studies.85 Predictions that target-of-rapamy- cyte–associated antigen 4 (CTLA-4) immune glob-
cin inhibitors plus alemtuzumab would induce tol- ulin that is a fusion protein combining CTLA-4
erance were also not confirmed. This combination (which engages CD80 and CD86) with the Fc por-
is associated with rejection episodes, including an- tion of IgG. Results of a phase 2 trial in patients un-
tibody-mediated rejection.86 Side effects of alemtu- dergoing renal transplantation who are receiving
zumab include first-dose reactions, neutropenia, mycophenolate mofetil, glucocorticoids, and anti-
anemia, and (rarely) pancytopenia and autoimmu- CD25 antibody are available in abstract form (www.
nity (e.g., hemolytic anemia, thrombocytopenia, atcmeeting.org/2004). In this trial with six months
and hyperthyroidism).87 The risks of immunodefi- of follow-up, the effect of repeated administration
ciency complications (infections and cancer) with of LEA29Y was similar to that of cyclosporine in
alemtuzumab are unknown. Alemtuzumab is used preventing rejection. The LEA29Y trial introduces
off-label for induction in some centers, but con- the concept of long-term use of nondepleting pro-

n engl j med 351;26 www.nejm.org december 23, 2004 2723

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

tein immunosuppressive agents to reduce reliance patients, rather than slowly deteriorating, as in the
on toxic small-molecule immunosuppressive drugs. past.57 This raises the hope that many organ trans-
plantations that are performed today represent a
additional drugs permanent cure for end-stage organ failure.
Many of the critical steps that are depicted in Fig- But concerns temper this optimism. Outcomes
ure 1 can be targeted by small molecules or proteins are not continuing to improve,103 and the rate of
to create new drugs.93 Potential targets for small- late graft loss remains excessive. For example, in
molecule drugs include those previously discussed the United States each year, end-stage kidney fail-
(e.g., calcineurin) as well as others (e.g., tyrosine ki- ure develops in 4500 patients who have undergone
nases, protein kinase Cθ, MAP kinases such as Jun kidney transplantation, a finding that highlights
N-terminal kinase, phosphoinositide-3-kinase, and transplant failure as a major cause of end-stage re-
chemokine receptors). Potential targets for protein nal disease.104 Patients who have undergone liver
drugs include many membrane proteins. transplantation have an excessive recurrence rate
Janus kinase 3 (JAK3) inhibitors39,40 illustrate of hepatitis; coronary artery disease develops in
how small-molecule immunosuppressive drugs some patients with transplanted hearts; and bron-
are developed. JAK3, a tyrosine kinase associated chiolitis obliterans often develops in patients with
with the cytokine receptor g chain, participates in transplanted lungs.105,106 The rate of premature
the signaling of many cytokine receptors (interleu- death with functioning allografts remains excessive,
kin-2, 4, 7, 9, 15, and 21) (Fig. 1). JAK3 inhibitor in part because of cardiovascular and other compli-
CP-690,55039 was developed by screening a chem- cations of immunosuppression.
ical library and modifying candidate compounds Nonimmune and immunodeficiency compli-
to produce an oral agent that is immunosuppres- cations of transplant immunosuppression should
sive in rodents and nonhuman primates. One ad- be reduced. The major nonimmune toxic effects are
verse effect is anemia, perhaps reflecting activity nephrotoxicity, hypertension, hyperlipidemia, dia-
against Janus kinase 2, which is needed for eryth- betes mellitus, and anemia. Five years after surgery,
ropoietin action. serious renal injury is present in 7 to 21 percent of
patients who have undergone nonrenal transplan-
tation,107 and end-stage kidney failure develops in
protocol development
and emerging issues many patients. The toxic effect of calcineurin in-
hibitors is an important contributor to the prob-
For two decades, the options for immunosuppres- lem of renal failure. Post-transplantation diabetes
sive drugs were initial induction with the use of mellitus develops after three years in 24 percent of
protein immunosuppressive therapy; preadapta- patients who have undergone renal transplanta-
tion maintenance therapy with three drugs — tion.108 Hyperlipidemia109 and anemia110 are com-
a calcineurin inhibitor, a second line of drugs (aza- mon and undertreated. Options for reducing toxic-
thioprine and now mycophenolate mofetil), and ity include choosing more selective drugs, avoiding
glucocorticoids; and postadaptation therapy with toxic combinations, and maintaining vigilance for
the same combination of drugs at lower doses. toxic effects.
Rejection was reversed with high-dose steroids or Cancers111 and infections that are induced by
depleting antibodies. Now hundreds of potential transplantation remain frequent, with infections
combinations exist, and many new protocols have now exceeding rejection in pediatric transplant re-
emerged, often including a reduced reliance on cipients.112 Choosing more selective drugs can re-
glucocorticoids94 and calcineurin inhibitors. Some duce these risks. For example, anti-CD25 antibody
examples are listed in Table 4. Developing evidence- has little effect on the risk of infection and post-
based approaches to this confusing choice of pro- transplantation lymphoproliferative disease.25 New
tocols presents a challenge. protocols must emphasize reducing the rates of
Progress in the control of early and late rejection cancer and infection rather than simply lowering
and in managing infections such as cytomegalovi- the rate of rejection.
rus has improved both the survival of patients and New immunosuppressive drug applications and
the function of grafts.57,100,101 For example, in kid- protocols113 are emerging without adequate trials
ney transplantation, the estimated glomerular fil- to establish dosing, safety, and efficacy. Examples
tration rate has improved102 and is stable in many are the regimens of induction with alemtuzumab or

2724 n engl j med 351;26 www.nejm.org december 23, 2004

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

Table 4. Examples of Current and Experimental Immunosuppressive Drug Protocols.

Protocol Protocol Elements Comments


Preadaptation Postadaptation
Protein Induction Maintenance Maintenance*
Conventional treatment Anti-CD25 antibody, Calcineurin inhibitor, my- Calcineurin inhibitor and Possibly excessive immunosup-
polyclonal antithy- cophenolate mofetil, mycophenolate mofetil; pression during postadaptation
mocyte globulin, and prednisone prednisone tapered
or none
Conventional treatment Anti-CD25 antibody Calcineurin inhibitor and Calcineurin inhibitor and Possible increase in rejection
with no steroids95 mycophenolate mofetil; mycophenolate mofetil;
prednisone only if prednisone only if
needed needed
Conventional treatment Polyclonal antithy- Calcineurin inhibitor, my- Calcineurin inhibitor and Effects of depletion (e.g., increased
with depleting anti- mocyte globulin cophenolate mofetil, mycophenolate mofetil; incidence of post-transplanta-
bodies and prednisone prednisone tapered tion lymphoproliferative dis-
order), possible late rejection
Sirolimus with cyclo- Anti-CD25 antibody, Cyclosporine, sirolimus, Sirolimus; prednisone Early toxicity of cyclosporine–siroli-
sporine withdrawal polyclonal antithy- and prednisone tapered mus combination
mocyte globulin,
or none
Calcineurin-inhibitor Anti-CD25 antibody, Sirolimus, mycophenolate Sirolimus and mycopheno- Possibly excessive early rejection;
avoidance with main- polyclonal antithy- mofetil, and prednisone late mofetil; prednisone no phase 3 trials; possible in-
tenance sirolimus mocyte globulin, tapered crease in late rejection
and mycophenolate or none
mofetil96,97
Calcineurin-inhibitor Anti-CD25 antibody, Calcineurin inhibitor, my- Mycophenolate mofetil; No phase 3 trials
withdrawal with my- polyclonal antithy- cophenolate mofetil, prednisone tapered
cophenolate mofetil mocyte globulin, and prednisone
maintenance98 or none
Alemtuzumab Alemtuzumab Sirolimus, prednisone Sirolimus; prednisone Long-term consequences of severe
induction84-86 tapered depletion unknown; no con-
trolled trials; possible increase
in antibody-mediated rejection
Depletion with minimi- Polyclonal antithy- Tacrolimus only if no Minimal tacrolimus if no Risk of late rejection as lymphoid
zation of immuno- mocyte globulin rejection rejection system recovers
suppressive drugs99
Maintenance with Anti-CD25 antibody CTLA-4–Ig, mycophenolate CTLA-4–Ig, mycophenolate Efficacy and safety must be estab-
CTLA-4–Ig and mofetil, and prednisone mofetil, and prednisone lished
mycophenolate
mofetil†

* For most protocols, no data are available regarding the relative cost and cost-effectiveness of the treatment and long-term requirements
for the administration of prednisone.
† CTLA-4–Ig denotes cytotoxic-T-lymphocyte–associated antigen 4 combined with the Fc portion of immunoglobulin G.

radical minimization of maintenance immunosup- function should not be dismissed as instances of


pression. Moreover, the quality of transplantation “chronic rejection”; instead, the source of injury
trials is suboptimal.114 One problem is that the should be diagnosed (e.g., rejection that is T-cell–
decline in the incidence of rejection, the end point mediated or antibody-mediated, recurrent disease,
in most trials, now limits the evaluation of new drug toxicity, or infection).116 The assumption must
agents.115 New composite end points could incor- be that new deterioration reflects new injury, not
porate organ function and drug toxicity or emerg- an inexorable consequence of an earlier injury. The
ing laboratory measurements of immune mecha- identification of mechanisms of injury may be re-
nisms. warded by the arresting of further deterioration.
Optimizing outcomes requires long-term fol- Robust tests for rejection that is T-cell–medi-
low-up by knowledgeable caregivers who recognize ated or antibody-mediated would change clinical
and react to changes. Allografts with deteriorating management and clinical trials (e.g., microarray

n engl j med 351;26 www.nejm.org december 23, 2004 2725

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

analysis of gene expression in biopsy specimens).11 ing true tolerance to HLA-incompatible organ trans-
Measurement of immune responses could guide plants is at hand. True tolerance is durable antigen-
transplantation management in the same way that specific unresponsiveness in an immunocompe-
measurement of disease activity guides other fields tent host that is induced by previous exposure to
(e.g., the measurement of lipid levels in the man- the antigen. The only clinical strategy that currently
agement of hyperlipidemia). meets this definition is stem-cell transplantation.122
Interest in suppressing alloantibody responses The stability of the adaptation usually depends on
is growing. Emerging evidence links alloantibody immunosuppression or damage to the immune
to late graft deterioration,19 and transplantation is tissues. At some point, most immunosuppressive
increasingly offered to patients who have previous- agents are billed as tolerogenic, an assertion that is
ly been excluded by existing alloantibody, includ- typically followed by the realization that, among
ing ABO blood-group barriers.117 Options include at least some patients, the immunologic tolerance
the optimization of baseline immunosuppression, is not durable after withdrawal of the drug therapy
the administration of rituximab or intravenous im- and recovery from its effects. Indeed, the first report
mune globulin, and plasmapheresis, but new strat- of an immunosuppressive drug was entitled “Drug-
egies are needed. Induced Immunological Tolerance.”123 Many “toler-
Pharmacogenomics offers possibilities for in- ance trials”124 in fact use immunosuppression and
dividualizing immunosuppression, an important are probably based on host–graft adaptation. Excel-
goal with respect to toxic drugs with narrow ther- lent immunosuppression with long-term clinical
apeutic indexes.118,119 For example, CYP3A (cyto- surveillance remains the best prospect for achiev-
chrome P-450-3A) allele CYP3A5*1, which is asso- ing the potential of transplantation to restore and
ciated with increased CYP3A5 levels, is present in maintain health.
70 to 80 percent of blacks but in only 5 to 10 per- Dr. Halloran reports having received lecture fees from Roche and
Fujisawa.
cent of whites.120 Since CYP3A5 genotyping can be I am indebted to Dr. Deborah James for her help in preparing the
used to predict slower achievement of target tacro- text and tables; to Drs. Karl Brinker, Arthur Matas, Sita Gourishan-
limus levels and earlier rejection,121 it could help kar, and Attapong Vongwiwatana for reviewing the manuscript; and
to Pam Publicover for her assistance in the preparation of the manu-
reduce rejection in black patients. script.
For most patients, no practical method of achiev-

refer enc es
1. Lombardi G, Sidhu S, Daly M, Batchelor 8. Biedermann BC, Pober JS. Human en- 14. Halloran PF, Urmson J, Ramassar V, et al.
JR, Makgoba W, Lechler RI. Are primary al- dothelial cells induce and regulate cytolytic Lesions of T-cell-mediated kidney allograft
loresponses truly primary? Int Immunol T cell differentiation. J Immunol 1998;161: rejection in mice do not require perforin or
1990;2:9-13. 4679-87. granzymes A and B. Am J Transplant 2004;
2. Adams AB, Williams MA, Jones TR, et 9. Wang D, Matsumoto R, You Y, et al. 4:705-12.
al. Heterologous immunity provides a potent CD3/CD28 costimulation-induced NF-kap- 15. Robertson H, Ali S, McDonnell BJ, Burt
barrier to transplantation tolerance. J Clin paB activation is mediated by recruitment of AD, Kirby JA. Chronic renal allograft dys-
Invest 2003;111:1887-95. protein kinase C-theta, Bcl10, and IkappaB function: the role of T cell-mediated tubular
3. Von Andrian UH, Mackay CR. T-cell func- kinase beta to the immunological synapse epithelial to mesenchymal cell transition.
tion and migration: two sides of the same through CARMA1. Mol Cell Biol 2004;24: J Am Soc Nephrol 2004;15:390-7.
coin. N Engl J Med 2000;343:1020-34. 164-71. 16. Halloran PF, Melk A, Barth C. Rethink-
4. Mandala S, Hajdu R, Bergstrom J, et al. 10. MacLennan IC, Toellner KM, Cunning- ing chronic allograft nephropathy: the con-
Alteration of lymphocyte trafficking by ham AF, et al. Extrafollicular antibody re- cept of accelerated senescence. J Am Soc
sphingosine-1-phosphate receptor agonists. sponses. Immunol Rev 2003;194:8-18. Nephrol 1999;10:167-81.
Science 2002;296:346-9. 11. Sarwal M, Chua M-S, Kambham N, et al. 17. Racusen LC, Colvin RB, Solez K, et al.
5. Bromley SK, Iaboni A, Davis SJ, et al. Molecular heterogeneity in acute renal allo- Antibody-mediated rejection criteria — an
The immunological synapse and CD28-CD80 graft rejection identified by DNA microarray addition to the Banff 97 classification of
interactions. Nat Immunol 2001;2:1159-66. profiling. N Engl J Med 2003;349:125-38. renal allograft rejection. Am J Transplant
6. Lakkis FG, Arakelov A, Konieczny BT, 12. Bogman MJ, Dooper IM, van de Winkel 2003;3:708-14.
Inoue Y. Immunologic ‘ignorance’ of vascu- JG, et al. Diagnosis of renal allograft rejec- 18. Halloran PF, Wadgymar A, Ritchie S,
larized organ transplants in the absence of tion by macrophage immunostaining with Falk J, Solez K, Srinivasa NS. The signifi-
secondary lymphoid tissue. Nat Med 2000; a CD14 monoclonal antibody, WT14. Lancet cance of the anti-class I antibody response. I.
6:686-8. 1989;2:235-8. Clinical and pathologic features of anti-class
7. Zhou P, Hwang KW, Palucki D, et al. Sec- 13. Rosenberg AS, Singer A. Cellular basis I-mediated rejection. Transplantation 1990;
ondary lymphoid organs are important but of skin allograft rejection: an in vivo model of 49:85-91.
not absolutely required for allograft respons- immune-mediated tissue destruction. Annu 19. Terasaki PI. Humoral theory of trans-
es. Am J Transplant 2003;3:259-66. Rev Immunol 1992;10:333-58. plantation. Am J Transplant 2003;3:665-73.

2726 n engl j med 351;26 www.nejm.org december 23, 2004

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

20. Feucht HE. Complement C4d in graft Sanders AG. Mycophenolic acid: an antibi- 50. Giblett ER, Anderson JE, Cohen F, Pol-
capillaries — the missing link in the recog- otic from Penicillium brevicompactum diercks. lara B, Meuwissen HJ. Adenosine-deami-
nition of humoral alloreactivity. Am J Trans- Lancet 1946;1:46-9. nase deficiency in two patients with severely
plant 2003;3:646-52. 37. Mitsui A, Suzuki S. Immunosuppres- impaired cellular immunity. Lancet 1972;2:
21. Starzl TE, Marchioro TL, Waddell WR. sive effect of mycophenolic acid. J Antibiot 1067-9.
The reversal of rejection in human renal ho- (Tokyo) 1969;22:358-63. 51. Allison AC, Hovi T, Watts RWE, Webster
mografts with subsequent development of 38. Brinkmann V, Lynch KR. FTY720: target- ADB. Immunological observations on pa-
homograft tolerance. Surg Gynecol Obstet ing G-protein-coupled receptors for sphin- tients with the Lesch-Nyhan syndrome, and
1963;117:385-95. gosine 1-phosphate in transplantation and on the role of de-novo purine synthesis in
22. Sakaguchi S. Naturally arising CD4+ autoimmunity. Curr Opin Immunol 2002;14: lymphocyte transformation. Lancet 1975;2:
regulatory T cells for immunologic self-tol- 569-75. 1179-83.
erance and negative control of immune re- 39. Changelian PS, Flanagan ME, Ball DJ, 52. Sollinger HW. Mycophenolate mofetil
sponses. Annu Rev Immunol 2004;22:531- et al. Prevention of organ allograft rejection for the prevention of acute rejection in pri-
62. by a specific Janus kinase 3 inhibitor. Science mary cadaveric renal allograft recipients.
23. Schwartz RH. T cell anergy. Annu Rev 2003;302:875-8. Transplant 1995;60:225-32.
Immunol 2003;21:305-34. 40. Behbod F, Erwin-Cohen RA, Wang M-E, 53. The Tricontinental Mycophenolate Mo-
24. Li Y, Li XC, Zheng XX, Wells AD, Turka et al. Concomitant inhibition of Janus ki- fetil Renal Transplantation Study Group. A
LA, Strom TB. Blocking both signal 1 and nase 3 and calcineurin-dependent signaling blinded, randomized clinical trial of myco-
signal 2 of T-cell activation prevents apopto- pathways synergistically prolongs the surviv- phenolate mofetil for the prevention of
sis of alloreactive T cells and induction of al of rat heart allografts. J Immunol 2001; acute rejection in cadaveric renal transplan-
peripheral allograft tolerance. Nat Med 166:3724-32. tation. Transplantation 1996;61:1029-37.
1999;5:1298-302. 41. Tiede I, Fritz G, Strand S, et al. CD28- 54. Halloran PF, Mathew T, Tomlanovich S,
25. Opelz G, Dohler B. Lymphomas after dependent Rac1 activation is the molecular Groth C, Hooftman L, Barker C. Mycophe-
solid organ transplantation: a Collaborative target of azathioprine in primary human nolate mofetil in renal allograft recipients:
Transplant Study report. Am J Transplant CD4+ T lymphocytes. J Clin Invest 2003; a pooled efficacy analysis of three random-
2004;4:222-30. 111:1133-45. ized, double-blind, clinical studies in preven-
26. Nickeleit V, Klimkait T, Binet IF, et al. 42. Clipstone NA, Crabtree GR. Identifica- tion of rejection. Transplant 1997;63:39-47.
Testing for polyomavirus type BK DNA in tion of calcineurin as a key signalling enzyme [Erratum, Transplantation 1997;63:618.]
plasma to identify renal-allograft recipients in T-lymphocyte activation. Nature 1992;357: 55. European Mycophenolate Mofetil Co-
with viral nephropathy. N Engl J Med 2000; 695-7. operative Study Group. Placebo-controlled
342:1309-15. 43. Clase CM, Mahalati K, Kiberd BA, et al. study of mycophenolate mofetil combined
27. Kazatchkine MD, Bellon B, Kaveri SV. Adequate early cyclosporin exposure is criti- with cyclosporin and corticosteroids for pre-
Mechanisms of action of intravenous immu- cal to prevent renal allograft rejection: pa- vention of acute rejection. Lancet 1995;345:
noglobulin (IVIG). Mult Scler 2000;6:Suppl tients monitored by absorption profiling. 1321-5.
2:S24-S26. Am J Transplant 2002;2:789-95. 56. Meier-Kriesche HU, Steffen BJ, Hoch-
28. Karin M. New twists in gene regulation 44. Cole E, Maham N, Cardella C, et al. Clin- berg AM, et al. Long-term use of mycophe-
by glucocorticoid receptor: is DNA binding ical benefits of neoral C2 monitoring in the nolate mofetil is associated with a reduction
dispensable? Cell 1998;93:487-90. long-term management of renal transplant in the incidence and risk of late rejection.
29. Batiuk TD, Pazderka F, Halloran PF. Cal- recipients. Transplantation 2003;75:2086- Am J Transplant 2003;3:68-73.
cineurin activity is only partially inhibited in 90. 57. Gourishankar S, Hunsicker LG, Jhangri
leukocytes of cyclosporine-treated patients. 45. Stalder M, Birsan T, Hubble RW, Pa- GS, Cockfield SM, Halloran PF. The stabili-
Transplantation 1995;59:1400-4. niagua RT, Morris RE. In vivo evaluation of ty of the glomerular filtration rate after renal
30. Elion GB. The George Hitchings and the novel calcineurin inhibitor ISATX247 in transplantation is improving. J Am Soc Neph-
Gertrude Elion Lecture: the pharmacology non-human primates. J Heart Lung Trans- rol 2003;14:2387-94.
of azathioprine. Ann N Y Acad Sci 1993;685: plant 2003;22:1343-52. 58. Kobashigawa J, Miller L, Renlund D,
400-7. 46. The U.S. Multicenter FK506 Liver Study et al. A randomized active-controlled trial
31. Borel JF, Feurer C, Gubler HJ, Stahelin Group. A comparison of tacrolimus (FK 506) of mycophenolate mofetil in heart transplant
H. Biological effects of cyclosporin A: a new and cyclosporine for immunosuppression recipients. Transplantation 1998;66:507-15.
antilymphocytic agent. Agents Actions 1976; in liver transplantation. N Engl J Med 1994; 59. Oz HS, Hughes WT. Novel anti-Pneu-
6:468-75. 331:1110-5. mocystis carinii effects of the immunosup-
32. Goto T, Kino T, Hatanaka H, et al. Dis- 47. European FK506 Multicentre Liver Study pressant mycophenolate mofetil in contrast
covery of FK-506, a novel immunosuppres- Group. Randomised trial comparing tacro- to provocative effects of tacrolimus, siroli-
sant isolated from streptomyces. J Am Chem limus (FK506) and cyclosporin in preven- mus, and dexamethasone. J Infect Dis 1997;
Soc 1987;109:5031-3. tion of liver allograft rejection. Lancet 1994; 175:901-4.
33. Kino T, Hatanaka H, Miyata S, et al. 344:423-8. 60. Gabardi S, Tran JL, Clarkson MR. Enter-
FK-506, a novel immunosuppressant isolat- 48. Ahsan N, Johnson C, Gonwa T, et al. ic-coated mycophenolate sodium. Ann Phar-
ed from a Streptomyces. II. Immunosup- Randomized trial of tacrolimus plus myco- macother 2003;37:1685-93.
pressive effect of FK-506 in vitro. J Antibiot phenolate mofetil or azathioprine versus 61. Vézina C, Kudelski A, Sehgal SN. Rapa-
(Tokyo) 1987;40:1256-65. cyclosporine oral solution (modified) plus mycin (AY-22,989), a new antifungal antibi-
34. Morris RE. Rapamycins: antifungal, an- mycophenolate mofetil after cadaveric kid- otic. I. Taxonomy of the producing strepto-
titumor, antiproliferative and immunosup- ney transplantation: results at 2 years. Trans- mycete and isolation of the active principle.
pressive macrolides. Transplant Rev 1992; plantation 2001;72:245-50. J Antibiot (Tokyo) 1975;28:721-6.
6:39-87. 49. Meier-Kriesche HU, Kaplan B. Cyclo- 62. Fritsche L, Budde K, Dragun D, Einecke
35. Gosio B. Ricerche batteriologiche e sporine microemulsion and tacrolimus are G, Diekmann F, Neumayer HH. Testosterone
chimiche sulle alterazion del mais; contrib- associated with decreased chronic allograft concentrations and sirolimus in male renal
uto alletiologia della pellagra. Riv Igiene San- failure and improved long-term graft sur- transplant patients. Am J Transplant 2004;
ita Pubblica Ann 1896;7:825-68. vival as compared with Sandimmune. Am 4:130-1.
36. Florey HW, Gilliver K, Jennings MA, J Transplant 2002;2:100-4. 63. Eisen HJ, Tuzcu EM, Dorent R, et al.

n engl j med 351;26 www.nejm.org december 23, 2004 2727

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
The new england journal of medicine

Everolimus for the prevention of allograft 77. Kahan BD, Karlix JL, Ferguson RM, et 91. Nashan B, Moore R, Amlot P, Schmidt
rejection and vasculopathy in cardiac-trans- al. Pharmacodynamics, pharmacokinetics, AG, Abeywickrama K, Soulillou JP. Ran-
plant recipients. N Engl J Med 2003;349:847- and safety of multiple doses of FTY720 in domised trial of basiliximab versus placebo
58. stable renal transplant patients: a multicenter, for control of acute cellular rejection in renal
64. MacDonald AS. A worldwide, phase III, randomized, placebo-controlled, phase I allograft recipients. Lancet 1997;350:1193-
randomized, controlled, safety and efficacy study. Transplantation 2003;76:1079-84. 8. [Erratum, Lancet 1997;350:1484.]
study of a sirolimus/cyclosporine regimen 78. Brennan DC, Flavin K, Lowell JA, et al. 92. Kahan BD, Rajagopalan PR, Hall M. Re-
for prevention of acute rejection in recipi- A randomized, double-blinded comparison duction of the occurrence of acute cellular
ents of primary mismatched renal allografts. of Thymoglobulin versus Atgam for induc- rejection among renal allograft recipients
Transplantation 2001;71:271-80. tion immunosuppressive therapy in adult re- treated with basiliximab, a chimeric anti-
65. Kahan BD. Efficacy of sirolimus com- nal transplant recipients. Transplantation interleukin-2-receptor monoclonal antibody.
pared with azathioprine for reduction of 1999;67:1011-8. Transplantation 1999;67:276-84.
acute renal allograft rejection: a randomised 79. Ortho Multicenter Transplant Study 93. Vincenti F. What’s in the pipeline? New
multicentre study. Lancet 2000;356:194- Group. A randomized clinical trial of OKT3 immunosuppressive drugs in transplanta-
202. monoclonal antibody for acute rejection of tion. Am J Transplant 2002;2:898-903.
66. McAlister VC, Gao Z, Peltekian K, cadaveric renal transplants. N Engl J Med 94. Hricik DE. Steroid-free immunosuppres-
Domingues J, Mahalati K, MacDonald AS. 1985;313:337-42. sion in kidney transplantation: an editorial
Sirolimus-tacrolimus combination immu- 80. Norman DJ, Kahana L, Stuart FP Jr, et al. review. Am J Transplant 2002;2:19-24.
nosuppression. Lancet 2000;355:376-7. A randomized clinical trial of induction 95. Cole E, Landsberg D, Russell D, et al.
67. Shapiro AMJ, Lakey JRT, Ryan EA, et al. therapy with OKT3 in kidney transplanta- A pilot study of steroid-free immunosup-
Islet transplantation in seven patients with tion. Transplantation 1993;55:44-50. pression in the prevention of acute rejection
type 1 diabetes mellitus using a glucocor- 81. Swinnen LJ, Costanzo-Nordin MR, Fish- in renal allograft recipients. Transplantation
ticoid-free immunosuppressive regimen. er SG, et al. Increased incidence of lympho- 2001;72:845-50.
N Engl J Med 2000;343:230-8. proliferative disorder after immunosuppres- 96. Kreis H, Cisterne JM, Land W, et al.
68. Gonwa T, Mendez R, Yang HC, Wein- sion with the monoclonal antibody OKT3 Sirolimus in association with mycopheno-
stein S, Jensik S, Steinberg S. Randomized in cardiac-transplant recipients. N Engl J Med late mofetil induction for the prevention of
trial of tacrolimus in combination with siroli- 1990;323:1723-8. acute graft rejection in renal allograft recipi-
mus or mycophenolate mofetil in kidney 82. Norman DJ, Vincenti F, de Mattos AM, ents. Transplantation 2000;69:1252-60.
transplantation: results at 6 months. Trans- et al. Phase I trial of HuM291, a humanized 97. Flechner SM, Goldfarb D, Modlin C, et al.
plant 2003;75:1213-20. anti-CD3 antibody, in patients receiving re- Kidney transplantation without calcineurin
69. Johnson RW, Kreis H, Oberbauer R, nal allografts from living donors. Transplan- inhibitor drugs: a prospective, randomized
Brattstrom C, Claesson K, Eris J. Sirolimus tation 2000;70:1707-12. trial of sirolimus versus cyclosporine. Trans-
allows early cyclosporine withdrawal in re- 83. Herold KC, Hagopian W, Auger JA, et al. plantation 2002;74:1070-6.
nal transplantation resulting in improved Anti-CD3 monoclonal antibody in new- 98. Smak Gregoor PJ, de Sevaux RG, Ligten-
renal function and lower blood pressure. onset type 1 diabetes mellitus. N Engl J Med berg G, et al. Withdrawal of cyclosporine or
Transplantation 2001;72:777-86. 2002;346:1692-8. prednisone six months after kidney trans-
70. Guba M, von Breitenbuch P, Steinbauer 84. Calne R, Friend P, Moffatt S, et al. Prope plantation in patients on triple drug therapy:
M, et al. Rapamycin inhibits primary and met- tolerance, perioperative campath 1H, and a randomized, prospective, multicenter study.
astatic tumor growth by antiangiogenesis: low-dose cyclosporin monotherapy in renal J Am Soc Nephrol 2002;13:1365-73.
involvement of vascular endothelial growth allograft recipients. Lancet 1998;351:1701- 99. Starzl TE, Murase N, Abu-Elmagd K, et
factor. Nat Med 2002;8:128-35. 2. [Erratum, Lancet 1998;352:408.] al. Tolerogenic immunosuppression for or-
71. Morice M-C, Serruys PW, Sousa JE, et al. 85. Kirk AD, Hale DA, Mannon RB, et al. gan transplantation. Lancet 2003;361:1502-
A randomized comparison of a sirolimus- Results from a human renal allograft toler- 10.
eluting stent with a standard stent for coro- ance trial evaluating the humanized CD52- 100. Hariharan S, Johnson CP, Bresnahan
nary revascularization. N Engl J Med 2002; specific monoclonal antibody alemtuzumab BA, Taranto SE, McIntosh MJ, Stablein D.
346:1773-80. (CAMPATH-1H). Transplantation 2003;76: Improved graft survival after renal trans-
72. Schwartz SM, Reidy MA, de Blois D. 120-9. plantation in the United States, 1988 to 1996.
Factors important in arterial narrowing. 86. Knechtle SJ, Pirsch JD, Fechner J Jr, et N Engl J Med 2000;342:605-12.
J Hypertens Suppl 1996;14:S71-S81. al. Campath-1H induction plus rapamycin 101. Port FK. Organ donation and trans-
73. Blum CB. Effects of sirolimus on lipids monotherapy for renal transplantation: re- plantation trends in the United States, 2001.
in renal allograft recipients: an analysis us- sults of a pilot study. Am J Transplant 2003; Am J Transplant 2003;3:Suppl 4:7-12.
ing the Framingham risk model. Am J Trans- 3:722-30. 102. Hariharan S, McBride MA, Cherikh WS,
plant 2002;2:551-9. 87. Coles AJ, Wing M, Smith S, et al. Pulsed Tolleris CB, Bresnahan BA, Johnson CP. Post-
74. Hardinger KL, Wang CD, Schnitzler MA, monoclonal antibody treatment and auto- transplant renal function in the first year pre-
et al. Prospective, pilot, open-label, short- immune thyroid disease in multiple sclero- dicts long-term kidney transplant survival.
term study of conversion to leflunomide re- sis. Lancet 1999;354:1691-5. Kidney Int 2002;62:311-8.
verses chronic renal allograft dysfunction. 88. Becker YT, Becker BN, Pirsch JD, 103. Meier-Kriesche HU, Schold JD, Srini-
Am J Transplant 2002;2:867-71. Sollinger HW. Rituximab as treatment for vas TR, Kaplan B. Lack of improvement in
75. Brinkmann V, Cyster JG, Hla T. FTY720: refractory kidney transplant rejection. Am renal allograft survival despite a marked de-
sphingosine 1-phosphate receptor-1 in the J Transplant 2004;4:996-1001. crease in acute rejection rates over the most
control of lymphocyte egress and endotheli- 89. Vincenti F, Kirkman R, Light S, et al. In- recent era. Am J Transplant 2004;4:378-83.
al barrier function. Am J Transplant 2004;4: terleukin-2-receptor blockade with daclizu- 104. U.S. Renal Data System. USRDS 2003
1019-25. mab to prevent acute rejection in renal trans- annual data report: atlas of end-stage renal
76. Budde K, Schmouder RL, Brunkhorst R, plantation. N Engl J Med 1998;338:161-5. disease in the United States. Bethesda, Md.:
et al. First human trial of FTY720, a novel 90. Nashan B, Light S, Hardie IR, Lin A, National Institutes of Health, National In-
immunomodulator, in stable renal trans- Johnson JR. Reduction of acute renal allo- stitutes of Diabetes & Digestive Kidney Dis-
plant patients. J Am Soc Nephrol 2002;13: graft rejection by daclizumab. Transplanta- eases, 2003.
1073-83. tion 1999;67:110-5. 105. Sharples LD, McNeil K, Stewart S, Wall-

2728 n engl j med 351;26 www.nejm.org december 23, 2004

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.
drug therapy

work J. Risk factors for bronchiolitis oblit- in the United States. Am J Transplant 2004; dividualization of drug administration. Am J
erans: a systematic review of recent publi- 4:905-13. Pharmacogenomics 2003;3:291-301.
cations. J Heart Lung Transplant 2002;21: 112. Dharnidharka VR, Stablein DM, Har- 119. Cattaneo D, Perico N, Remuzzi G. From
271-81. mon WE. Post-transplant infections now pharmacokinetics to pharmacogenomics:
106. Stewart KC, Patterson GA. Current exceed acute rejection as cause for hospi- a new approach to tailor immunosuppressive
trends in lung transplantation. Am J Trans- talization: a report of the NAPRTCS. Am therapy. Am J Transplant 2004;4:299-310.
plant 2001;1:204-10. J Transplant 2004;4:384-9. 120. MacPhee IA, Fredericks S, Tai T, et al.
107. Ojo AO, Held PJ, Port FK, et al. Chronic 113. Matas AJ. What’s new and what’s hot Tacrolimus pharmacogenetics: polymor-
renal failure after transplantation of a non- in transplantation: clinical science ATC 2003. phisms associated with expression of cyto-
renal organ. N Engl J Med 2003;349:931-40. Am J Transplant 2003;3:1465-73. chrome p4503A5 and P-glycoprotein corre-
108. Kasiske BL, Snyder JJ, Gilbertson D, 114. Fritsche L, Einecke G, Fleiner F, Dra- late with dose requirement. Transplantation
Matas AJ. Diabetes mellitus after kidney gun D, Neumayer HH, Budde K. Reports of 2002;74:1486-9.
transplantation in the United States. Am large immunosuppression trials in kidney 121. Idem. The influence of pharmacogenet-
J Transplant 2003;3:178-85. transplantation: room for improvement. Am ics on the time to achieve target tacrolimus
109. Kasiske B, Cosio FG, Beto J, et al. Clin- J Transplant 2004;4:738-43. concentrations after kidney transplantation.
ical practice guidelines for managing dyslip- 115. Hariharan S, McBride MA, Cohen EP. Am J Transplant 2004;4:914-9.
idemias in kidney transplant patients: a re- Evolution of endpoints for renal transplant 122. Auchincloss H Jr. In search of the elu-
port from the Managing Dyslipidemias in outcome. Am J Transplant 2003;3:933-41. sive holy grail: the mechanisms and pros-
Chronic Kidney Disease Work Group of the 116. Halloran PF. Call for revolution: a new pects for achieving clinical transplantation
National Kidney Foundation Kidney Disease approach to describing allograft deteriora- tolerance. Am J Transplant 2001;1:6-12.
Outcomes Quality Initiative. Am J Trans- tion. Am J Transplant 2002;2:195-200. 123. Schwartz R, Dameshek W. Drug-
plant 2004;4:Suppl 7:13-53. 117. Warren DS, Zachary AA, Sonnenday CJ, induced immunological tolerance. Nature
110. Vanrenterghem Y, Ponticelli C, Mo- et al. Successful renal transplantation across 1959;183:1682-3.
rales JM, et al. Prevalence and management simultaneous ABO incompatible and posi- 124. Matthews JB, Ramos E, Bluestone JA.
of anemia in renal transplant recipients: tive crossmatch barriers. Am J Transplant Clinical trials of transplant tolerance: slow
a European survey. Am J Transplant 2003;3: 2004;4:561-8. but steady progress. Am J Transplant 2003;
835-45. 118. Fredericks S, Holt DW, MacPhee IA. 3:794-803.
111. Kasiske BL, Snyder JJ, Gilbertson DT, The pharmacogenetics of immunosuppres- Copyright © 2004 Massachusetts Medical Society.
Wang C. Cancer after kidney transplantation sion for organ transplantation: a route to in-

journal editorial fellow


The Journal’s editorial office invites applications for a one-year
research fellowship beginning in July 2005 from individuals at any
stage of training. The editorial fellow will work on Journal projects
and will participate in the day-to-day editorial activities of the
Journal but is expected in addition to have his or her own inde-
pendent projects. Please send curriculum vitae and research in-
terests to the Editor-in-Chief, 10 Shattuck St., Boston, MA 02115
(fax, 617-739-9864), by January 15, 2005.

n engl j med 351;26 www.nejm.org december 23, 2004 2729

The New England Journal of Medicine


Downloaded from nejm.org at UNIV OF UTAH ECCLES on July 5, 2014. For personal use only. No other uses without permission.
Copyright © 2004 Massachusetts Medical Society. All rights reserved.

You might also like