Liver Transplantation in Adults: Overview of Immunosuppression
Liver Transplantation in Adults: Overview of Immunosuppression
Liver Transplantation in Adults: Overview of Immunosuppression
All topics are updated as new evidence becomes available and our peer review process is complete.
Literature review current through: Feb 2023. | This topic last updated: Sep 24, 2021.
INTRODUCTION
In 1994, the US Multicenter FK506 Liver Study Group published a paper comparing cyclosporine
and tacrolimus for immunosuppression after liver transplantation [1]. The study was a
landmark in the evolution of liver transplantation. First, the introduction stated that rejection
remained an important cause of graft loss and death. Second, the paper reported that survival
with cyclosporine and tacrolimus was similar but that tacrolimus was associated with fewer
episodes of steroid-resistant rejection. Third, it reported that tacrolimus was associated with
excess adverse events, including nephrotoxicity and neurotoxicity.
Liver transplantation has evolved substantially since that publication, and we have addressed
many of the issues outlined in the 1994 study. Acute rejection is usually easy to manage, and we
now try to balance the risk of rejection with the risk of drug toxicity. Our focus has shifted to
avoiding the long-term complications of immunosuppression and recurrent liver disease.
Tacrolimus has become first-line immunosuppression in most liver transplant programs, and a
number of supplemental drugs allow us to customize immunosuppression.
This topic will provide an overview of the drugs used for immunosuppression following liver
transplantation. The diagnosis and treatment of acute T-cell mediated (cellular) rejection are
discussed elsewhere. (See "Liver transplantation in adults: Clinical manifestations and diagnosis
of acute T-cell mediated (cellular) rejection of the liver allograft" and "Liver transplantation in
adults: Treatment of acute T cell-mediated (cellular) rejection of the liver allograft".)
https://www.uptodate.com/contents/4586/print 1/39
3/29/23, 7:58 PM 4586
Signal III: Clonal expansion — Newly synthesized IL-2 is secreted by T cells and binds to IL-2
receptors (IL-2R) on the cell surface in an autocrine fashion, stimulating a burst of cell
proliferation. Basiliximab and daclizumab, both monoclonal antibodies against the IL-2
receptor, block this signal. Sirolimus, which binds to the downstream mechanistic target of
rapamycin (mTOR), also acts at this step. Finally, the proliferation burst can be inhibited at the
level of DNA synthesis by azathioprine and mycophenolate mofetil. (See 'Basiliximab' below and
'Sirolimus' below and 'Inhibitors of purine and pyrimidine synthesis' below.)
DRUG INTERACTIONS
https://www.uptodate.com/contents/4586/print 2/39
3/29/23, 7:58 PM 4586
A major issue with the immunosuppressive agents used for liver transplant recipients
(particularly calcineurin inhibitors and mechanistic target of rapamycin inhibitors) is their
extensive metabolism by CYP3A4. This creates the potential for drug-drug interactions that may
produce toxicity or dangerously low levels of immunosuppressive agents, leading to an
increased risk of rejection. As examples, antifungal agents, some antibiotics, and many of the
drugs used in the treatment of HIV inhibit CYP3A4. (See "Kidney transplantation in adults:
Kidney transplantation in patients with HIV".)
A knowledge of the common interfering compounds is essential and drug levels must be
monitored closely when these compounds are used ( table 1).
GLUCOCORTICOIDS
Steroids are associated with a number of side effects including diabetes, fluid retention,
hypertension, emotional lability, hyperlipidemia, cosmetic changes (acne, buffalo hump, etc),
poor wound healing, susceptibility to infection, visual changes, cataracts, and osteopenia, and
they may cause adrenal suppression that may persist up to the time of weaning [5].
In addition to the side effects, steroid therapy increases hepatitis C virus (HCV) replication. The
basis of this increase is controversial. Steroids may drive replication directly, and/or they may
permit more effective replication through immunosuppression. In vitro studies using a replicon
system (which avoids the effect of the immune system) have yielded conflicting results [6,7].
https://www.uptodate.com/contents/4586/print 3/39
3/29/23, 7:58 PM 4586
The controversy about steroid avoidance and tapering in HCV-infected patients is discussed
below, but it is mostly of historical interest with the availability of potent direct acting antiviral
agents. (See 'Patients with HCV infection' below.)
Given the problems with glucocorticoids, many centers try to wean from steroids as early as
possible. This must be done cautiously since rapid steroid withdrawal can precipitate a flare of
an underlying condition (eg, autoimmune hepatitis, inflammatory bowel disease, or hepatitis)
or an episode of rejection. A meta-analysis looked at 16 randomized trials that compared
postoperative glucocorticoid avoidance or withdrawal with glucocorticoid-containing
immunosuppression [8]. There were no differences detected in mortality, graft loss, or infection
rates when postoperative glucocorticoid avoidance or withdrawal was compared with
glucocorticoid-containing immunosuppression. However, acute rejection and glucocorticoid-
resistant rejection were more common with glucocorticoid avoidance or withdrawal (relative
risk [RR] 1.33, 95% CI 1.08 to 1.64, and RR 2.14, 95% CI 1.13 to 4.02, respectively). Diabetes
mellitus and hypertension occurred less often with glucocorticoid avoidance or withdrawal (RR
0.81, 95% CI 0.66 to 0.99 and RR 0.76, 95% CI 0.65 to 0.90, respectively). However, all of the
studies included in the analysis were at high risk of bias.
Patients with HCV infection — A great deal of literature had been devoted to optimizing
immunosuppression for patients who were transplanted for HCV-related liver disease [12-22].
However, the availability of safe and effective HCV therapy with direct-acting antivirals has
revolutionized the approach to HCV management in liver transplant candidates and recipients.
Many patients are successfully treated for HCV infection before liver transplantation.
https://www.uptodate.com/contents/4586/print 4/39
3/29/23, 7:58 PM 4586
CALCINEURIN INHIBITORS
The early challenges to successful liver transplantation included surgical technique, organ
preservation, and immunosuppression. As surgical technique improved, the need for better
immunosuppression became more obvious. A report published in 1980 described one-year
survival of only 26 percent [23]. The introduction of the calcineurin inhibitor (CNI) cyclosporine A
the following year marked a turning point in liver transplantation [24,25].
The impact of cyclosporine was illustrated in an early report in which one- and five-year survival
rates with "conventional" immunosuppression (azathioprine plus prednisone) were 33 and 20
percent, respectively, while the survival rates with cyclosporine plus prednisone were 70 and 63
percent, respectively [27]. Cyclosporine subsequently became first-line therapy, and the first
1000 cases done at the University of Pittsburgh were described in a report in 1988 [28].
After oral administration, cyclosporine is variably absorbed in the jejunum and enters the
lymphatic system. Peak blood levels are achieved in two to four hours, and the drug is widely
https://www.uptodate.com/contents/4586/print 5/39
3/29/23, 7:58 PM 4586
distributed with highest concentrations in adipose, pancreatic, adrenal, renal, and hepatic
tissues. The average half-life is 15 hours but ranges widely (10 to 40 hours). Cyclosporine is
cleared in the bile after extensive metabolism in the liver by CYP3A4. The metabolites have little
immunosuppressive activity. CYP3A4 activity (and, therefore, blood cyclosporine levels) depends
upon genetic and environmental factors including gene polymorphisms, graft function,
hepatitis C virus (HCV) replication, and certain foods and drugs ( table 1).
Cyclosporine levels should be monitored frequently in the peritransplant period (typically daily),
with decreasing frequency as graft function stabilizes and rejection becomes less of a threat.
Patients who are stable can be monitored monthly, but levels should be checked more
frequently if they develop an acute illness or start taking a potentially interfering drug. Several
drugs commonly affect CNI levels. The goal therapeutic level of cyclosporine is usually 200 to
250 ng/mL in the first three months after transplantation, but is typically tapered to 80 to 120
ng/mL by 12 months.
Cyclosporine levels should be monitored closely, and patients should be monitored for renal
toxicity, hypertension, hyperkalemia, and hypomagnesemia. Potassium-sparing diuretics and
potentially nephrotoxic drugs should be avoided if possible. Neurologic toxicity may include
altered mental status, polyneuropathy, dysarthria, myoclonus, seizures, hallucinations, and
cortical blindness [29]. Other common problems include hyperlipidemia, gingival hyperplasia,
and hirsutism. (See "Pharmacology of cyclosporine and tacrolimus".)
Cyclosporine versus tacrolimus — By the mid-1990s, most centers agreed that tacrolimus was
associated with superior graft and patient survival. A criticism of the early studies was their use
of oil-based cyclosporine, raising the question of bioavailability versus efficacy. A landmark
study greatly influenced thinking on the subject [32]. The authors compared the efficacy of
tacrolimus versus microemulsified cyclosporine in 606 patients undergoing first orthotopic liver
https://www.uptodate.com/contents/4586/print 6/39
3/29/23, 7:58 PM 4586
Both treatment regimens were effective, but tacrolimus was superior with regard to the
composite endpoint and for patient and graft survival ( figure 3). In addition, more patients in
the tacrolimus group survived without an episode of significant rejection.
The results of the study were updated with a two-year extension of the randomization protocol
[33]. Tacrolimus remained superior for the composite endpoint (RR 0.79; 95% CI 0.60-0.95),
although the individual endpoints of freedom from death or retransplantation were no longer
statistically significant. Significantly more patients randomized to tacrolimus were alive with
their original graft and on their assigned medication compared with the cyclosporine group (62
versus 42 percent). Six patients were switched from tacrolimus to cyclosporine, while 17 were
switched from cyclosporine to tacrolimus (five for graft rejection not meeting endpoint criteria).
Multiple subsequent studies have been performed; the superiority of tacrolimus over
cyclosporine was confirmed in a meta-analysis of 16 randomized clinical trials [34,35].
Tacrolimus was superior when analyzed for survival, graft loss, acute rejection, and steroid-
resistant rejection in the first year ( table 5). The incidence of lymphoproliferative disease was
similar for the two groups, and de novo diabetes mellitus was more common in the tacrolimus
group. More patients stopped cyclosporine than tacrolimus. The authors estimated that
treating 100 patients with tacrolimus versus cyclosporine would avoid rejection, steroid-
resistant rejection, graft loss, and death in nine, seven, five, and two patients, respectively, but
that four additional patients would develop diabetes.
Tacrolimus dosing — Tacrolimus dosing should be individualized. We usually start with a low
dose (0.5 to 1 mg every 12 hours) on postoperative day 1, and aim for a level of 7 to 10 ng/mL
by the end of the first week. We use lower dosing, often with the addition of an auxiliary agent
like mycophenolate mofetil (MMF) or a monoclonal antibody in patients with preoperative renal
impairment. It is important to attain adequate tacrolimus levels quickly. In a study of 493 liver
transplantation recipients who were treated with tacrolimus as their primary
immunosuppression, patients with trough tacrolimus levels >7 ng/mL at the time of a protocol
https://www.uptodate.com/contents/4586/print 7/39
3/29/23, 7:58 PM 4586
liver biopsy (mean 6 days after transplantation) had lower rates of moderate or severe rejection
compared with those who had lower levels (24 versus 41 percent) [36]. In addition, patients with
mean levels between 7 and 10 ng/mL within 15 days after liver transplantation had lower rates
of graft loss during follow-up compared with patients who had trough levels <7 ng/mL or 10 to
15 ng/mL (relative risks of 2.32 and 2.17, respectively). These findings suggest that in the early
post-transplantation period, a tacrolimus level between 7 and 10 ng/mL is associated with
improved outcomes. However, it must be kept in mind that because the study was not a
randomized trial, it is at risk for bias and confounding.
A level of 6 ng/mL is usually satisfactory after six months, and maintenance at a level of 4 to 6
ng/mL is common beyond one year. We aim for higher levels in patients who are transplanted
for autoimmune liver diseases, including primary biliary cholangitis (PBC) and primary
sclerosing cholangitis. Patients transplanted for alcohol-associated liver disease or
hemochromatosis usually tolerate low levels of CNIs after their initial recovery. With improved
survival, our goal is to use as little immunosuppression as possible to minimize the known long-
term complications of these drugs, including renal insufficiency and post-transplant
lymphoproliferative disorders (PTLD).
Calcineurin inhibitors and renal failure — CNI-induced renal failure is a serious problem after
orthotopic liver transplant (OLT) [37]. The problem has been exacerbated by the switch to a
MELD-based organ allocation system, which is weighted towards higher serum creatinine. (See
"Model for End-stage Liver Disease (MELD)".)
A prospective, multicenter trial compared standard dose tacrolimus to low dose and delayed
tacrolimus with the primary endpoint of a change in eGFR (Cockroft-Gault formula) at 52 weeks
[39]. Patients were divided into three groups:
● Group A (n = 183): standard-dose tacrolimus (target trough >10 ng/mL) and steroids
● Group B (n = 70): MMF 2 g/day, low-dose tacrolimus (target trough 8 ng/mL), and steroids
https://www.uptodate.com/contents/4586/print 8/39
3/29/23, 7:58 PM 4586
● Group C (n = 172): daclizumab induction, MMF, reduced-dose tacrolimus delayed until the
fifth day post-transplant, and steroids
The eGFR decreased by 23.6, 21.2, and 13.6 mL/min in groups A, B, and C, respectively (A versus
C, p = 0.012; A versus B, p = 0.199).
Hemodialysis was required more frequently in group A compared with group C (10 versus 4
percent). Biopsy-proven acute rejection rates were 28, 29, and 19 percent, respectively. Patient
and graft survival were similar among the groups. This study suggests that the kidney is
particularly vulnerable to injury in the immediate post-OLT period. Delayed CNI dosing reduced,
but did not eliminate, renal injury.
Patients with HCV infection — The availability of highly effective antiviral agents (DAAs) has
greatly simplified post-OLT HCV therapy. Antiviral agents may interfere with drug metabolism
because of effects on CYP3A4 and/or P-glycoprotein (gp) [40-45]. In addition, the rate of
calcineurin inhibitor clearance may increase with a declining viral load. For these reasons,
calcineurin inhibitor levels should be monitored closely after starting DAAs. (See "Hepatitis C
virus infection in liver transplant candidates and recipients", section on 'Interactions with
immunosuppressive agents'.)
Drug interactions can also be checked through the Lexicomp drug interactions program
included with UpToDate.
Summary on the role of calcineurin inhibitors — Cyclosporine and tacrolimus are potent
immunosuppressive agents. Their availability has allowed us to shift our focus from acute
cellular rejection and short-term post-transplant survival to long-term management of
complications. They have similar adverse effects including nephrotoxicity, neurotoxicity, and
electrolyte abnormalities, and both can be monitored with drug levels.
https://www.uptodate.com/contents/4586/print 9/39
3/29/23, 7:58 PM 4586
The effectiveness of sirolimus was illustrated in an early report of its use in 15 patients
undergoing liver transplantation [47]. Sirolimus was used as a single agent or as part of dual
(sirolimus and cyclosporine) or triple therapy (sirolimus, cyclosporine, and prednisolone).
Rejection was seen more commonly with monotherapy, rarely with dual therapy, and not at all
with triple therapy. In addition, all patients were on sirolimus monotherapy by three months.
Only 3 of the 15 patients discontinued sirolimus: one for hyperlipidemia, one for taste
perversion, and one for Pneumocystis pneumonia.
Although sirolimus binds its target (FK-binding protein) with higher affinity than tacrolimus, the
two drugs act synergistically, rather than competitively, to prevent rejection. This led to
speculation that a low-dose combination strategy might reduce the incidence of tacrolimus-
associated problems. In a study of 56 patients on this combination, patient and graft survival at
23 months were 93 and 91 percent, respectively [48]. One case of hepatic artery thrombosis was
observed in this series.
A retrospective analysis showed no benefit for renal function when patients with chronic renal
insufficiency were switched from CNIs to sirolimus [54]. This study suggests that renal
https://www.uptodate.com/contents/4586/print 10/39
3/29/23, 7:58 PM 4586
dysfunction from CNIs becomes irreversible at some point, and highlights the importance of
CNI optimization. Similarly, another study showed that early conversion (less than 90 days) to
sirolimus was associated with improved renal function, while late conversion was of limited
benefit [55].
A systematic review that included 11 studies found a small (3.4 mL/min), nonsignificant increase
in glomerular filtration rate after one year of sirolimus use in patients who received sirolimus as
primary immunosuppression due to renal insufficiency or who were switched to sirolimus from
another regimen due to nephrotoxicity [56]. However, sirolimus use was associated with higher
rates of infection, rash, ulcers, and discontinuation of therapy. Sirolimus was not associated
with an increased risk of graft failure or death, though the data reporting for these outcomes
were incomplete.
Larger trials with longer follow-up are needed to settle the issue and to determine whether the
potential improvement in renal function outweighs a possible increased risk of rejection.
Sirolimus has also been proposed as a better choice for patients with hepatocellular carcinoma
because of its antiproliferative activity [57-59]. This benefit has yet to be proven in prospective
trials. Finally, although not supported by publications, many transplant centers find that
sirolimus is inadequate as monotherapy and routinely add a second agent when switching from
a CNI for any reason.
Side effects — Side effects of sirolimus reported in the postoperative period include hepatic
artery thrombosis, delayed wound healing, and incisional hernias, while chronic use has been
associated with hyperlipidemia, bone marrow suppression, mouth ulcers, skin rashes,
albuminuria, and pneumonia. These risks are difficult to quantify because the incidence (and
even the presence) of side effects varies widely by report. In 2008, the FDA updated the labeling
of sirolimus to include a boxed warning stating that the use of sirolimus was associated with
excess mortality, graft loss, and hepatic artery thrombosis following liver transplantation, and
that its use de novo in liver transplantation recipients was not recommended [60].
As an example, one report showed that sirolimus was more likely to cause hyperlipidemia when
administered with cyclosporine than with tacrolimus (30 versus 6 percent for
hypercholesterolemia, 33 versus 3 percent for hypertriglyceridemia) [61]. Similar results in
another retrospective trial suggested that the combination of sirolimus with tacrolimus is
associated with minimal elevation of triglycerides [62]. These contrast with a study that
reported a 49 percent incidence of hyperlipidemia in patients switched from a CNI to sirolimus
monotherapy [63].
https://www.uptodate.com/contents/4586/print 11/39
3/29/23, 7:58 PM 4586
Another retrospective study compared 170 patients treated with sirolimus as initial therapy
compared with 180 historic controls [64]. No significant differences in wound complications (12
versus 14 percent) or hepatic artery complications (5 versus 8 percent) were seen [64]. Finally, a
large retrospective study found complications that included edema, dermatitis, oral ulcers, joint
pains, pleural effusions, hepatic artery thrombosis (two patients), and one wound dehiscence
[65]. The results reported in the above studies have not been validated in prospective studies.
An open-label randomized trial found that patients who were switched from a CNI to sirolimus
had increased rates of acute rejection but similar mortality at 12 months [66].
The initial immunosuppressive regimen after transplantation and optimal timing of withdrawal
of CNI is not clear [68]. Three trials of post-transplant regimens including EVR compared with
standard CNI therapy have generally shown benefit in renal function parameters for the EVR
groups but its efficacy compared with standard CNI therapy needs further study [68-70]. (See
'Calcineurin inhibitors and renal failure' above.)
● In a trial of 188 liver transplant recipients, all of whom initially received basiliximab
induction therapy and enteric coated mycophenolate sodium (with or without steroids),
renal function was better after 24 weeks in patients receiving EVR and tacrolimus
(EVR+TAC) while tapering tacrolimus (TAC) with discontinuation by week 16, compared with
patients receiving continuous TAC (mean eGFR 95.8 versus 76.0 mL/min) [68]. Rates of
treatment failure (defined as biopsy-proven acute rejection, graft loss or death) at 24
weeks were not significantly different between the EVR+TAC and the TAC groups.
● In a trial of 303 liver transplant recipients with GFR >50 mL/min who received basiliximab
induction therapy followed by CNI (with or without steroids) for four weeks post-
transplantation and who then continued CNI or were converted to EVR, the mean
calculated GFR (Cockroft-Gault) at 11 months showed no difference between regimens
(-2.9 mL/min in favor of EVR, 95% CI [-10.65 to 4.81]) [69]. The difference in GFR was
significant if the Modification of Diet in Renal Disease formula was used (-7.8 mL/min in
favor of EVR; 95% CI: -14.366 to -1.191). Rates of mortality and biopsy-proven acute
rejection were similar in both groups. (See "Assessment of kidney function".)
https://www.uptodate.com/contents/4586/print 12/39
3/29/23, 7:58 PM 4586
Everolimus is the hydroxyethyl derivative of sirolimus. The mechanism of action of EVR is via
inhibition of mammalian target of rapamycin (mTOR), similar to sirolimus [71]. Everolimus is
rapidly absorbed and reaches a peak concentration within one to two hours if given on an
empty stomach [72]. Fatty foods retard absorption. It has higher oral availability and lower
plasma binding than sirolimus and a mean elimination half-life of 30±11 hours. A starting dose
of 0.75 mg twice daily with a target trough level of 3 to 8 ng/mL is standard [73]. Metabolism is
via CYP3A4, 3A5, and 2C8, and interactions with azoles, macrolides, antiepileptic agents,
antiviral agents, and grapefruit juice are known. The clearance of everolimus is about 20
percent higher in Black patients.
Side effects — Side effects of everolimus seem to be dose related and are similar to those
seen for sirolimus, [72] and may include anemia, peripheral edema, elevations in serum
creatinine when used with full dose CNIs, diarrhea, nausea, urinary tract infections, and
hyperlipidemia. Mouth ulcers were not seen in heart and kidney transplantation trials, but they
were reported in the liver transplantation trials.
The major antimetabolite drugs are prodrugs of mycophenolic acid (MPA), a purine synthesis
inhibitor, and azathioprine.
The original drugs in this class, cyclophosphamide (Cytoxan) and azathioprine (Imuran), are
rarely used in transplantation in the United States, although a multicenter study suggested that
azathioprine is used routinely in the United Kingdom [19]. While mycophenolate mofetil (MMF)
has become more popular than the other agents, clear evidence of superiority is lacking [74].
https://www.uptodate.com/contents/4586/print 13/39
3/29/23, 7:58 PM 4586
Azathioprine is sometimes substituted for MMF in women who are pregnant or of childbearing
age due to increased safety experience with it in pregnancy. MMF is pregnancy class D.
Reports on the use of MMF in liver transplantation began to appear in the late 1990s [76-78].
MMF does not cause neurotoxicity or nephrotoxicity, and is widely used as a calcineurin
inhibitor (CNI)- or steroid-sparing agent. The most common side effects are bone marrow
suppression and gastrointestinal complaints, including abdominal pain, ileus, nausea, vomiting,
and oral ulceration. These symptoms are usually dose-related and improve with temporary or
permanent dose reduction. Usual dosing is 1 g twice daily. Patients may tolerate the drug better
if it is initially dosed at 500 mg twice daily or 500 mg four times daily. Myfortic (mycophenolate
sodium) is formulated as 360 mg tablets and is typically given as two tablets (720 mg) orally
every 12 hours. Food may interfere with absorption of the drugs, so they should be taken one
hour before or two hours after meals.
The role of MMF is similar to that of sirolimus in that it is used to reduce or discontinue CNI
dosing in order to treat side effects. Studies suggest MMF monotherapy may be effective in
certain situations long after liver transplantation:
● A randomized trial assigned 150 patients who had received a liver transplantation and
were maintained on a CNI to either continued therapy with a CNI or to MMF monotherapy
[79]. The mean time between liver transplantation and study enrollment was 4.9 years for
patients assigned to continued CNI therapy, and 5.7 years for those assigned to MMF.
After five years of follow-up, there were no significant differences between those who
continued on a CNI and those who were switched to MMF with regard to chronic rejection
or patient survival (0 versus 0 percent and 94 versus 90 percent, respectively). There was a
https://www.uptodate.com/contents/4586/print 14/39
3/29/23, 7:58 PM 4586
trend toward lower acute rejection rates in the CNI group (3 versus 11 percent, p = 0.055).
All patients with acute rejection were successfully treated with steroid-pulse therapy.
Among patients with renal insufficiency, renal function improved in those switched to
MMF. There were no significant differences between the groups with regard to malignancy
rates or to adverse cardiovascular, gastrointestinal, or neurologic events.
The added immunosuppression of MMF may also allow for the early discontinuation or
avoidance of steroids [81].
Side effects of azathioprine include bone marrow suppression, nausea, vomiting, pancreatitis,
hepatotoxicity, and neoplasia. (See "Pharmacology and side effects of azathioprine when used
in rheumatic diseases", section on 'Adverse effects'.)
ANTIBODY THERAPY
Because T and B cells express specific antigens on their cell surfaces, antibodies to these
markers can be used to deplete populations of cells. The initial concept of lymphoid depletion
by thoracic duct fistula [83] gave way to the more elegant method of antithymocyte globulin
preparations, now refined to include humanized antibodies and monoclonal antibodies against
specific cell surface proteins such as the interleukin (IL)-2 receptor [84].
https://www.uptodate.com/contents/4586/print 15/39
3/29/23, 7:58 PM 4586
These agents are not used routinely in liver transplantation, but have an important role in
treating steroid-resistant rejection and as calcineurin inhibitor (CNI)-sparing agents in the
immediate post-transplant period. Antibody therapy may also permit steroid free
immunosuppression regimens if needed. We generally use antibody preparations in the rare
case of steroid-resistant rejection and in situations where we wish to minimize CNI dosing, such
as in patients with pretransplant renal failure. (See "Liver transplantation in adults: Treatment of
acute T cell-mediated (cellular) rejection of the liver allograft", section on 'Therapy for
nonresponders'.)
In addition, a large series examined the efficacy of rabbit ATG following liver transplantation
[87]. The series included 500 patients who received a single dose of solumedrol followed by ATG
induction. Patients also received mycophenolate mofetil and tacrolimus or sirolimus. The
tacrolimus or sirolimus was weaned after three months. After one year, patient and graft
survival rates were 93 and 90 percent, respectively. Rejection occurred in 114 patients (23
percent) and 33 patients required glucocorticoids (7 percent).
Complications with these agents include fever, chills, rash, anemia, thrombocytopenia, serum
sickness, and nephritis. Although our personal preference is to use monoclonal antibodies
when necessary, some reports suggest that polyclonal antibodies are still in use in pediatric and
adult patients undergoing liver transplantation [89,90].
Monoclonal antibodies
(OKT3, no longer available), and they have longer half-lives and are better tolerated.
Basiliximab, for example, has an elimination half-life of 4.1±2.1 days, and complete saturation
of interleukin-2 receptor alpha-chain was maintained as long as serum concentrations
exceeded 0.1 microgram/mL [91]. The mean duration of receptor saturation was 23 +/- 7 days
after transplantation (range of 13 to 41 days). The half-life may be decreased by the presence of
bleeding or ascites.
Similar efficacy has been demonstrated with daclizumab dosed at 2 mg/kg on post-orthotopic
liver transplant (OLT) days 1 and 3, and 1 mg/kg on day 8 [92]. CD25 suppression was confirmed
through day 30, and maximal effects were noted with a daclizumab concentration of at least 5
micrograms/mL. Similarly, a nonrandomized study of daclizumab 2 mg/kg before engraftment
and 1 mg/kg on day 5 post-OLT resulted in a much lower rejection rate in the first six months
(18 versus 40 percent), marked improvement in renal function, and no increase in
cytomegalovirus (CMV) or infectious complications when compared with a control group treated
with standard immunosuppression [93].
However, daclizumab was removed from the market in 2018 due to safety concerns following
several reports of inflammatory encephalitis and meningoencephalitis [94,95].
Antibodies can be used to reduce CNI use in patients with pre-OLT renal disease [96] or to
minimize steroid use [22,97]. In one report with median follow-up of 22 months, basiliximab
controlled steroid-resistant rejection after OLT in five out of seven children [98].
INVESTIGATIONAL AGENTS
Belatacept (LEA29Y) — Belatacept is a high affinity fusion protein that binds CD80/86 on
antigen-presenting cells (APCs). This prevents binding of CD80/86 to CD28 on the T cell (signal II
in Figure 1) ( figure 1), and blocks the costimulatory pathway [100]. The drug is given as a
https://www.uptodate.com/contents/4586/print 17/39
3/29/23, 7:58 PM 4586
monthly infusion and may permit immunosuppression without nephrotoxicity. Several reports
confirm its effectiveness in renal transplantation [101-103], but an increased rate of post-
transplant lymphoproliferative disorder (especially involving the central nervous system [104])
has confined its use to Epstein-Barr virus-positive recipients. We are unaware of belatacept use
in liver transplantation.
A few other agents, including alefacept and a Janus tyrosine kinase 3 inhibitor, are in early
testing in renal transplantation [105].
Patients should continue to take their immunosuppressive medication around the time of
surgery, although the route of administration may need to be changed if the patient cannot
tolerate oral medication. Intravenous administration of calcineurin inhibitors (CNIs) is
associated with a higher risk of nephrotoxicity and should be avoided if possible. In addition,
clinicians should be careful to check for potential drug interactions with medications used
around the time of surgery (eg, antimicrobials or antifungals). The patient's transplant
hepatologist should be consulted prior to making any changes to the immunosuppression
regimen. (See 'Drug interactions' above.)
https://www.uptodate.com/contents/4586/print 18/39
3/29/23, 7:58 PM 4586
STOPPING IMMUNOSUPPRESSION
Some patients will develop immunologic tolerance following liver transplantation and may be
able to stop taking immunosuppressants [110]. However, because of the risk of graft rejection
and because it is not yet clear which patients are good candidates for stopping
immunosuppression, additional studies are needed before recommending attempts at
stopping immunosuppression. We have seen a number of fatal rejections in patients who
stopped immunosuppression without medical supervision, including patients many years post-
orthotopic liver transplant. We therefore never recommend stopping all immunosuppression.
Links to society and government-sponsored guidelines from selected countries and regions
around the world are provided separately. (See "Society guideline links: Liver transplantation".)
https://www.uptodate.com/contents/4586/print 19/39
3/29/23, 7:58 PM 4586
● For patients with pretransplant renal failure in whom we wish to minimize the use of
calcineurin inhibitors (CNI), we generally use antibody preparations in the immediate post-
transplant period along with delayed calcineurin inhibitors. (See 'Antibody therapy' above
and 'Calcineurin inhibitors' above.)
● Slowly worsening renal disease in the late post-orthotopic liver transplant period can be
managed by reducing the CNI dose, with the addition of MMF, or by switching to sirolimus
or everolimus. (See 'Inhibitors of mammalian (mechanistic) target of rapamycin (mTOR)'
above.)
● Some patients will develop immunologic tolerance following liver transplantation and may
be able to stop taking immunosuppressants. However, because of the risk of irreversible
graft rejection, and because we have no tools to assess which patients are good
candidates for stopping immunosuppression, we never recommend complete cessation of
immunosuppression. (See 'Stopping immunosuppression' above.)
ACKNOWLEDGMENT
The UpToDate editorial staff acknowledges Norman Sussman, MD, who contributed to an
earlier version of this topic review.
REFERENCES
1. U.S. Multicenter FK506 Liver Study Group. A comparison of tacrolimus (FK 506) and
cyclosporine for immunosuppression in liver transplantation. N Engl J Med 1994; 331:1110.
2. Vacca A, Felli MP, Farina AR, et al. Glucocorticoid receptor-mediated suppression of the
interleukin 2 gene expression through impairment of the cooperativity between nuclear
factor of activated T cells and AP-1 enhancer elements. J Exp Med 1992; 175:637.
https://www.uptodate.com/contents/4586/print 20/39
3/29/23, 7:58 PM 4586
3. Ray A, LaForge KS, Sehgal PB. On the mechanism for efficient repression of the interleukin-
6 promoter by glucocorticoids: enhancer, TATA box, and RNA start site (Inr motif) occlusion.
Mol Cell Biol 1990; 10:5736.
4. Verhoef CM, van Roon JA, Vianen ME, et al. The immune suppressive effect of
dexamethasone in rheumatoid arthritis is accompanied by upregulation of interleukin 10
and by differential changes in interferon gamma and interleukin 4 production. Ann Rheum
Dis 1999; 58:49.
5. Toniutto P, Fabris C, Fumolo E, et al. Prevalence and risk factors for delayed adrenal
insufficiency after liver transplantation. Liver Transpl 2008; 14:1014.
6. Henry SD, Metselaar HJ, Van Dijck J, et al. Impact of steroids on hepatitis C virus replication
in vivo and in vitro. Ann N Y Acad Sci 2007; 1110:439.
7. Kim SS, Peng LF, Lin W, et al. A cell-based, high-throughput screen for small molecule
regulators of hepatitis C virus replication. Gastroenterology 2007; 132:311.
8. Fairfield C, Penninga L, Powell J, et al. Glucocorticosteroid-free versus glucocorticosteroid-
containing immunosuppression for liver transplanted patients. Cochrane Database Syst
Rev 2018; 4:CD007606.
9. Manns MP, Woynarowski M, Kreisel W, et al. Budesonide induces remission more effectively
than prednisone in a controlled trial of patients with autoimmune hepatitis.
Gastroenterology 2010; 139:1198.
10. Bhat M, Ghali P, Wong P, et al. Immunosuppression with budesonide for liver transplant
recipients with severe infections. Liver Transpl 2012; 18:262.
11. Bari K, Shah SA, Kaiser TE, et al. Safety and Efficacy of Budesonide for Liver Transplant
Immune Suppression: Results of a Pilot Phase 2a Trial. Liver Transpl 2020; 26:1430.
12. Vivarelli M, Burra P, La Barba G, et al. Influence of steroids on HCV recurrence after liver
transplantation: A prospective study. J Hepatol 2007; 47:793.
13. Klintmalm GB, Davis GL, Teperman L, et al. A randomized, multicenter study comparing
steroid-free immunosuppression and standard immunosuppression for liver transplant
recipients with chronic hepatitis C. Liver Transpl 2011; 17:1394.
14. Lladó L, Xiol X, Figueras J, et al. Immunosuppression without steroids in liver
transplantation is safe and reduces infection and metabolic complications: results from a
prospective multicenter randomized study. J Hepatol 2006; 44:710.
15. Eason JD, Nair S, Cohen AJ, et al. Steroid-free liver transplantation using rabbit
antithymocyte globulin and early tacrolimus monotherapy. Transplantation 2003; 75:1396.
https://www.uptodate.com/contents/4586/print 21/39
3/29/23, 7:58 PM 4586
16. Marcos A, Eghtesad B, Fung JJ, et al. Use of alemtuzumab and tacrolimus monotherapy for
cadaveric liver transplantation: with particular reference to hepatitis C virus.
Transplantation 2004; 78:966.
17. Berenguer M, Aguilera V, Prieto M, et al. Significant improvement in the outcome of HCV-
infected transplant recipients by avoiding rapid steroid tapering and potent induction
immunosuppression. J Hepatol 2006; 44:717.
18. Lladó L, Fabregat J, Castellote J, et al. Impact of immunosuppression without steroids on
rejection and hepatitis C virus evolution after liver transplantation: results of a prospective
randomized study. Liver Transpl 2008; 14:1752.
19. Manousou P, Cholongitas E, Samonakis D, et al. Reduced fibrosis in recurrent HCV with
tacrolimus, azathioprine and steroids versus tacrolimus: randomised trial long term
outcomes. Gut 2014; 63:1005.
20. Takada Y, Kaido T, Asonuma K, et al. Randomized, multicenter trial comparing tacrolimus
plus mycophenolate mofetil to tacrolimus plus steroids in hepatitis C virus-positive
recipients of living donor liver transplantation. Liver Transpl 2013; 19:896.
21. Sgourakis G, Radtke A, Fouzas I, et al. Corticosteroid-free immunosuppression in liver
transplantation: a meta-analysis and meta-regression of outcomes. Transpl Int 2009;
22:892.
22. Washburn K, Speeg KV, Esterl R, et al. Steroid elimination 24 hours after liver
transplantation using daclizumab, tacrolimus, and mycophenolate mofetil. Transplantation
2001; 72:1675.
23. Starzl TE, Koep L, Porter KA, et al. Decline in survival after liver transplantation. Arch Surg
1980; 115:815.
24. Starzl TE, Klintmalm GB, Porter KA, et al. Liver transplantation with use of cyclosporin a and
prednisone. N Engl J Med 1981; 305:266.
25. Calne RY. Liver transplantation. Ann Clin Res 1981; 13:327.
26. Borel JF, Feurer C, Gubler HU, Stähelin H. Biological effects of cyclosporin A: a new
antilymphocytic agent. Agents Actions 1976; 6:468.
27. Gordon RD, Shaw BW Jr, Iwatsuki S, et al. Indications for liver transplantation in the
cyclosporine era. Surg Clin North Am 1986; 66:541.
28. Iwatsuki S, Starzl TE, Todo S, et al. Experience in 1,000 liver transplants under cyclosporine-
steroid therapy: a survival report. Transplant Proc 1988; 20:498.
https://www.uptodate.com/contents/4586/print 22/39
3/29/23, 7:58 PM 4586
30. Gordon RD, Starzl TE. Changing perspectives on liver transplantation in 1988. Clin Transpl
1988; :5.
31. Starzl TE, Todo S, Fung J, et al. FK 506 for liver, kidney, and pancreas transplantation. Lancet
1989; 2:1000.
32. O'Grady JG, Burroughs A, Hardy P, et al. Tacrolimus versus microemulsified ciclosporin in
liver transplantation: the TMC randomised controlled trial. Lancet 2002; 360:1119.
33. O'Grady JG, Hardy P, Burroughs AK, et al. Randomized controlled trial of tacrolimus versus
microemulsified cyclosporin (TMC) in liver transplantation: poststudy surveillance to 3
years. Am J Transplant 2007; 7:137.
34. McAlister VC, Haddad E, Renouf E, et al. Cyclosporin versus tacrolimus as primary
immunosuppressant after liver transplantation: a meta-analysis. Am J Transplant 2006;
6:1578.
35. Haddad EM, McAlister VC, Renouf E, et al. Cyclosporin versus tacrolimus for liver
transplanted patients. Cochrane Database Syst Rev 2006; :CD005161.
36. Rodríguez-Perálvarez M, Germani G, Papastergiou V, et al. Early tacrolimus exposure after
liver transplantation: relationship with moderate/severe acute rejection and long-term
outcome. J Hepatol 2013; 58:262.
37. Ojo AO, Held PJ, Port FK, et al. Chronic renal failure after transplantation of a nonrenal
organ. N Engl J Med 2003; 349:931.
38. Lake J, Patel D, David K, et al. The association between MMF and risk of progressive renal
dysfunction and death in adult liver transplant recipients with HCV. Clin Transplant 2009;
23:108.
39. Neuberger JM, Mamelok RD, Neuhaus P, et al. Delayed introduction of reduced-dose
tacrolimus, and renal function in liver transplantation: the 'ReSpECT' study. Am J Transplant
2009; 9:327.
40. Martin P, Busuttil RW, Goldstein RM, et al. Impact of tacrolimus versus cyclosporine in
hepatitis C virus-infected liver transplant recipients on recurrent hepatitis: a prospective,
randomized trial. Liver Transpl 2004; 10:1258.
41. Rayhill SC, Barbeito R, Katz D, et al. A cyclosporine-based immunosuppressive regimen may
be better than tacrolimus for long-term liver allograft survival in recipients transplanted for
hepatitis C. Transplant Proc 2006; 38:3625.
42. Berenguer M, Royuela A, Zamora J. Immunosuppression with calcineurin inhibitors with
respect to the outcome of HCV recurrence after liver transplantation: results of a meta-
analysis. Liver Transpl 2007; 13:21.
https://www.uptodate.com/contents/4586/print 23/39
3/29/23, 7:58 PM 4586
47. Watson CJ, Friend PJ, Jamieson NV, et al. Sirolimus: a potent new immunosuppressant for
liver transplantation. Transplantation 1999; 67:505.
48. McAlister VC, Peltekian KM, Malatjalian DA, et al. Orthotopic liver transplantation using low-
dose tacrolimus and sirolimus. Liver Transpl 2001; 7:701.
49. Neff GW, Montalbano M, Tzakis AG. Ten years of sirolimus therapy in orthotopic liver
transplant recipients. Transplant Proc 2003; 35:209S.
50. Beckebaum S, Cicinnati V, Brokalaki E, et al. CNI-sparing regimens within the liver
transplant setting: experiences of a single center. Clin Transpl 2004; :215.
51. Watson CJ, Gimson AE, Alexander GJ, et al. A randomized controlled trial of late conversion
from calcineurin inhibitor (CNI)-based to sirolimus-based immunosuppression in liver
transplant recipients with impaired renal function. Liver Transpl 2007; 13:1694.
52. Campbell MS, Rai J, Kozin E, et al. Effects of sirolimus vs. calcineurin inhibitors on renal
dysfunction after orthotopic liver transplantation. Clin Transplant 2007; 21:377.
53. Shenoy S, Hardinger KL, Crippin J, et al. Sirolimus conversion in liver transplant recipients
with renal dysfunction: a prospective, randomized, single-center trial. Transplantation
2007; 83:1389.
54. DuBay D, Smith RJ, Qiu KG, et al. Sirolimus in liver transplant recipients with renal
dysfunction offers no advantage over low-dose calcineurin inhibitor regimens. Liver
Transpl 2008; 14:651.
55. Rogers CC, Johnson SR, Mandelbrot DA, et al. Timing of sirolimus conversion influences
recovery of renal function in liver transplant recipients. Clin Transplant 2009; 23:887.
56. Asrani SK, Leise MD, West CP, et al. Use of sirolimus in liver transplant recipients with renal
insufficiency: a systematic review and meta-analysis. Hepatology 2010; 52:1360.
https://www.uptodate.com/contents/4586/print 24/39
3/29/23, 7:58 PM 4586
58. Toso C, Meeberg GA, Bigam DL, et al. De novo sirolimus-based immunosuppression after
liver transplantation for hepatocellular carcinoma: long-term outcomes and side effects.
Transplantation 2007; 83:1162.
59. Zimmerman MA, Trotter JF, Wachs M, et al. Sirolimus-based immunosuppression following
liver transplantation for hepatocellular carcinoma. Liver Transpl 2008; 14:633.
60. http://www.accessdata.fda.gov/drugsatfda_docs/label/2008/021083s033,021110s043lbl.pdf
(Accessed on April 17, 2013).
61. Trotter JF, Wachs ME, Trouillot TE, et al. Dyslipidemia during sirolimus therapy in liver
transplant recipients occurs with concomitant cyclosporine but not tacrolimus. Liver
Transpl 2001; 7:401.
62. Kniepeiss D, Iberer F, Schaffellner S, et al. Dyslipidemia during sirolimus therapy in patients
after liver transplantation. Clin Transplant 2004; 18:642.
63. Morard I, Dumortier J, Spahr L, et al. Conversion to sirolimus-based immunosuppression in
maintenance liver transplantation patients. Liver Transpl 2007; 13:658.
64. Dunkelberg JC, Trotter JF, Wachs M, et al. Sirolimus as primary immunosuppression in liver
transplantation is not associated with hepatic artery or wound complications. Liver Transpl
2003; 9:463.
65. Montalbano M, Neff GW, Yamashiki N, et al. A retrospective review of liver transplant
patients treated with sirolimus from a single center: an analysis of sirolimus-related
complications. Transplantation 2004; 78:264.
66. Abdelmalek MF, Humar A, Stickel F, et al. Sirolimus conversion regimen versus continued
calcineurin inhibitors in liver allograft recipients: a randomized trial. Am J Transplant 2012;
12:694.
67. http://www.fda.gov/Safety/MedWatch/SafetyInformation/ucm303659.htm (Accessed on Ma
rch 12, 2013).
68. Saliba F, Duvoux C, Gugenheim J, et al. Efficacy and Safety of Everolimus and Mycophenolic
Acid With Early Tacrolimus Withdrawal After Liver Transplantation: A Multicenter
Randomized Trial. Am J Transplant 2017; 17:1843.
69. Fischer L, Klempnauer J, Beckebaum S, et al. A randomized, controlled study to assess the
conversion from calcineurin-inhibitors to everolimus after liver transplantation--PROTECT.
Am J Transplant 2012; 12:1855.
https://www.uptodate.com/contents/4586/print 25/39
3/29/23, 7:58 PM 4586
70. De Simone P, Nevens F, De Carlis L, et al. Everolimus with reduced tacrolimus improves
renal function in de novo liver transplant recipients: a randomized controlled trial. Am J
Transplant 2012; 12:3008.
71. Levy G, Schmidli H, Punch J, et al. Safety, tolerability, and efficacy of everolimus in de novo
liver transplant recipients: 12- and 36-month results. Liver Transpl 2006; 12:1640.
73. Shipkova M, Hesselink DA, Holt DW, et al. Therapeutic Drug Monitoring of Everolimus: A
Consensus Report. Ther Drug Monit 2016; 38:143.
74. Germani G, Pleguezuelo M, Villamil F, et al. Azathioprine in liver transplantation: a
reevaluation of its use and a comparison with mycophenolate mofetil. Am J Transplant
2009; 9:1725.
75. Everson GT. Everolimus and mTOR inhibitors in liver transplantation: opening the "box".
Liver Transpl 2006; 12:1571.
76. Hood KA, Zarembski DG. Mycophenolate mofetil: a unique immunosuppressive agent. Am J
Health Syst Pharm 1997; 54:285.
77. Stegall MD, Wachs ME, Everson G, et al. Prednisone withdrawal 14 days after liver
transplantation with mycophenolate: a prospective trial of cyclosporine and tacrolimus.
Transplantation 1997; 64:1755.
78. Klupp J, Bechstein WO, Platz KP, et al. Mycophenolate mofetil added to immunosuppression
after liver transplantation--first results. Transpl Int 1997; 10:223.
79. Schmeding M, Kiessling A, Neuhaus R, et al. Mycophenolate mofetil monotherapy in liver
transplantation: 5-year follow-up of a prospective randomized trial. Transplantation 2011;
92:923.
80. Hebert MF, Ascher NL, Lake JR, et al. Four-year follow-up of mycophenolate mofetil for graft
rescue in liver allograft recipients. Transplantation 1999; 67:707.
81. O'Grady JG. Corticosteroid-free strategies in liver transplantation. Drugs 2006; 66:1853.
82. Perry I, Neuberger J. Immunosuppression: towards a logical approach in liver
transplantation. Clin Exp Immunol 2005; 139:2.
83. Starzl TE, Koep LJ, Halgrimson CG, et al. Liver transplantation--1978. Transplant Proc 1979;
11:240.
84. Briggs JD. A critical review of immunosuppressive therapy. Immunol Lett 1991; 29:89.
85. Eason JD, Loss GE, Blazek J, et al. Steroid-free liver transplantation using rabbit
antithymocyte globulin induction: results of a prospective randomized trial. Liver Transpl
https://www.uptodate.com/contents/4586/print 26/39
3/29/23, 7:58 PM 4586
2001; 7:693.
86. Penninga L, Wettergren A, Wilson CH, et al. Antibody induction versus placebo, no
induction, or another type of antibody induction for liver transplant recipients. Cochrane
Database Syst Rev 2014; :CD010253.
87. Yoo MC, Vanatta JM, Modanlou KA, et al. Steroid-free Liver Transplantation Using Rabbit
Antithymocyte Globulin Induction in 500 Consecutive Patients. Transplantation 2015;
99:1231.
88. Shapiro R, Young JB, Milford EL, et al. Immunosuppression: evolution in practice and
trends, 1993-2003. Am J Transplant 2005; 5:874.
89. Shah A, Agarwal A, Mangus R, et al. Induction immunosuppression with rabbit
antithymocyte globulin in pediatric liver transplantation. Liver Transpl 2006; 12:1210.
90. Varo E, López A, Rivero C. Initial immunosuppression in liver transplant recipients with
impaired renal function. Transplant Proc 2005; 37:3909.
91. Kovarik J, Breidenbach T, Gerbeau C, et al. Disposition and immunodynamics of basiliximab
in liver allograft recipients. Clin Pharmacol Ther 1998; 64:66.
92. Washburn WK, Teperman LW, Heffron TG, et al. A novel three-dose regimen of daclizumab
in liver transplant recipients with hepatitis C: a pharmacokinetic and pharmacodynamic
study. Liver Transpl 2006; 12:585.
93. Eckhoff DE, McGuire B, Sellers M, et al. The safety and efficacy of a two-dose daclizumab
(zenapax) induction therapy in liver transplant recipients. Transplantation 2000; 69:1867.
94. http://www.ema.europa.eu/ema/index.jsp?curl=pages/news_and_events/news/2018/03/ne
ws_detail_002917.jsp&mid=WC0b01ac058004d5c1 (Accessed on March 05, 2018).
95. http://media.biogen.com/press-release/autoimmune-diseases/biogen%C2%A0and-abbvie-
announce%C2%A0-voluntary%C2%A0worldwide-withdrawal-marketi (Accessed on March 0
5, 2018).
96. Emre S, Gondolesi G, Polat K, et al. Use of daclizumab as initial immunosuppression in liver
transplant recipients with impaired renal function. Liver Transpl 2001; 7:220.
97. Liu CL, Fan ST, Lo CM, et al. Interleukin-2 receptor antibody (basiliximab) for
immunosuppressive induction therapy after liver transplantation: a protocol with early
elimination of steroids and reduction of tacrolimus dosage. Liver Transpl 2004; 10:728.
98. Aw MM, Taylor RM, Verma A, et al. Basiliximab (Simulect) for the treatment of steroid-
resistant rejection in pediatric liver transpland recipients: a preliminary experience.
Transplantation 2003; 75:796.
https://www.uptodate.com/contents/4586/print 27/39
3/29/23, 7:58 PM 4586
107. Alcaide ML, Abbo L, Pano JR, et al. Herpes zoster infection after liver transplantation in
patients receiving induction therapy with alemtuzumab. Clin Transplant 2008; 22:502.
108. Silveira FP, Husain S, Kwak EJ, et al. Cryptococcosis in liver and kidney transplant recipients
receiving anti-thymocyte globulin or alemtuzumab. Transpl Infect Dis 2007; 9:22.
109. Dhesi S, Boland B, Colquhoun S. Alemtuzumab and liver transplantation: a review. Curr
Opin Organ Transplant 2009; 14:245.
110. Levitsky J. Operational tolerance: past lessons and future prospects. Liver Transpl 2011;
17:222.
111. de la Garza RG, Sarobe P, Merino J, et al. Trial of complete weaning from
immunosuppression for liver transplant recipients: factors predictive of tolerance. Liver
Transpl 2013; 19:937.
Topic 4586 Version 37.0
https://www.uptodate.com/contents/4586/print 28/39
3/29/23, 7:58 PM 4586
GRAPHICS
Signal III: Newly synthesized IL-2 and growth factors feed back on T cell membrane
receptors, causing clonal expansion of newly activated T cells.
Reproduced with permission from: Vierling, JM. Clinical Use of Immunosuppressive Drugs to Control the
Immune Response. In: Liver Immunology: Principles and Practice, Gershwin, ME, Vierling, JM, Manns,
MP (Eds), Humana Press, Totowa 2007. Copyright ©2007 Springer-Verlag.
https://www.uptodate.com/contents/4586/print 29/39
3/29/23, 7:58 PM 4586
Reproduced with permission from: Vierling, JM. Clinical Use of Immunosuppressive Drugs to Control the
Immune Response. In: Liver Immunology: Principles and Practice, Gershwin, ME, Vierling, JM, Manns, MP (Eds),
Humana Press, Totowa 2007. Copyright ©2007 Springer-Verlag.
https://www.uptodate.com/contents/4586/print 30/39
3/29/23, 7:58 PM 4586
Approach to management in
Common types of
Examples of interacting drugs the absence of appropriate
drug interactions
noninteracting alternatives
https://www.uptodate.com/contents/4586/print 31/39
3/29/23, 7:58 PM 4586
https://www.uptodate.com/contents/4586/print 32/39
3/29/23, 7:58 PM 4586
Important note: The interactions listed in this table illustrate some of the common types of
interactions with immunosuppressive drugs; this is not a complete list, and many other
significant drug interactions can occur.
Cyclosporine, tacrolimus, sirolimus, and everolimus are highly dependent upon CYP3A
metabolism for clearance and are also substrates of P-gp drug efflux pump. Some interactions
can lead to subtherapeutic or dangerously toxic levels of immunosuppressant concentrations.
When appropriate noninteracting alternatives are readily available, consider modifying treatment
to avoid combined use with potent metabolic inhibitors/inducers or agents known to have
additive toxicities with immunosuppressants.
Drug therapy should be managed by transplant specialists with expertise in therapeutic drug
monitoring, and doses should be adjusted based upon measurement of immunosuppressant
concentrations, particularly whenever drug therapy is altered. If there are any concerns about
the safety of a given medication or supplement, they should be discussed with the patient's
transplant center prior to initiation.
CYP: cytochrome P450 metabolism; P-gp: P-glycoprotein drug efflux pump; ART: HIV antiretroviral
therapy; HIV: human immunodeficiency virus; HCV: hepatitis C virus; ACE: angiotensin-converting
enzyme; ARB: angiotensin II receptor blocker.
Prepared with data from Lexicomp Online. Copyright © 1978-2023 Lexicomp, Inc. All Rights Reserved.
https://www.uptodate.com/contents/4586/print 33/39
3/29/23, 7:58 PM 4586
Hydrocortisone 20
Prednisolone 5
Prednisone 5
Methylprednisolone 4
https://www.uptodate.com/contents/4586/print 34/39
3/29/23, 7:58 PM 4586
https://www.uptodate.com/contents/4586/print 35/39
3/29/23, 7:58 PM 4586
Cyclosporine Twice daily Capsules, suspension, Drug level, creatinine, lipids, K(+),
parenteral Mg(2+), CNS toxicity
https://www.uptodate.com/contents/4586/print 36/39
3/29/23, 7:58 PM 4586
Reproduced with permission from: O'Grady, JG, et al. Tacrolimus versus microemulsified
ciclosporin in liver transplantation: the TMC randomised controlled trial. Lancet 2002; 360:1119.
Copyright ©2002 Elsevier.
https://www.uptodate.com/contents/4586/print 37/39
3/29/23, 7:58 PM 4586
Data from: McAlister, VC, Haddad, E, Renouf, E, et al. Cyclosporine versus tacrolimus as primary immunosuppressant after
liver transplantation: a meta-analysis. Am J Transplant 2006; 6:1578.
https://www.uptodate.com/contents/4586/print 38/39
3/29/23, 7:58 PM 4586
Contributor Disclosures
John M Vierling, MD, FACP Equity Ownership/Stock Options: Athenex, Inc [Oncology Therapeutics].
Grant/Research/Clinical Trial Support: Allergan [NASH]; Enanta [NASH]; Gilead [PBC, PSC, NASH]; Intercept
[PBC, NASH]; Lilly [PBC]; Novartis [PBC, AIH, NASH, OLT]; Pliant [PSC]; Taiwan J [AIH]. Consultant/Advisory
Boards: Arena [AIH, PBC, PSC]; Blade [PSC, NASH]; Enanta [NASH]; Gilead [PBC, PSC]; Intercept [PBC,
NASH]; Kezar [AIH]; Lilly [PBC]; Moderna [AIH, AILDs]; Novartis [PBC, AIH, NASH, OLT]; Perspectum [Liver
Multiplex imaging, MRCP-Plus imaging]; Selecta [PBC, AIH, PSC]; Taiwan J [AIH]. Speaker's Bureau: Chronic
Liver Disease Foundation [CME only]. Other Financial Interest: Athenex Inc – Biopharmaceutical company
developing novel oncology therapies, including CAR-NKT and CAR-T cell technologies [Member, Board of
Directors]. All of the relevant financial relationships listed have been mitigated. Robert S Brown, Jr, MD,
MPH Grant/Research/Clinical Trial Support: AbbVie [Hepatitis C]; DURECT [Alcoholic hepatitis]; Enanta
[Nonalcoholic fatty liver disease]; Genfit [Nonalcoholic fatty liver disease]; Gilead [Hepatitis C]; Intercept
[Nonalcoholic fatty liver disease]; Mallinckrodt [Hepatorenal, Hepatitis C]; Mirum [Pruritus, cholestasis];
Salix [Encephalopathy, hepatorenal, Hepatitis C, nonalcoholic fatty liver disease]. Consultant/Advisory
Boards: AbbVie [Hepatitis B, hepatitis C, primary biliary cholangitis, cirrhosis]; Ambys [Hepatocyte
transplantation]; Antios [Hepatitis B]; Gilead [Hepatitis B, hepatitis C, hepatitis D]; Intercept [NASH,
primary biliary cholangitis, cirrhosis]; Mallinckrodt [Hepatorenal syndrome]; Mirum [Pruritus, cholestasis];
Salix [Hepatic encephalopathy]. All of the relevant financial relationships listed have been
mitigated. Kristen M Robson, MD, MBA, FACG No relevant financial relationship(s) with ineligible
companies to disclose.
Contributor disclosures are reviewed for conflicts of interest by the editorial group. When found, these are
addressed by vetting through a multi-level review process, and through requirements for references to be
provided to support the content. Appropriately referenced content is required of all authors and must
conform to UpToDate standards of evidence.
https://www.uptodate.com/contents/4586/print 39/39