You are on page 1of 10

Transplantation Reviews xxx (2016) xxxxxx

Contents lists available at ScienceDirect

Transplantation Reviews
journal homepage: www.elsevier.com/locate/trre

Use of everolimus in liver transplantation: The French experience


Jrme Dumortier a, Sebastien Dharancy b, Yvon Calmus c, Christophe Duvoux d, Franois Durand e,
Ephrem Salam f, Faouzi Saliba g,
a
Unit de Transplantation Hpatique-Fdration des Spcialits Digestives, Hpital Edouard Herriot and Universit Claude Bernard Lyon 1, Lyon, France
b
Service des Maladies de l'Appareil Digestif et de la Nutrition, Hpital Claude Huriez Centre Hospitalier Universitaire, Lille, France
c
Unit Mdicale de Transplantation Hpatique, AP-HP Hpital Universitaire Piti Salptrire, Paris, France
d
Service d'Hpato-Gastroenterologie, Hpital Henri Mondor, Assistance Publique, Hpitaux de Paris, Crteil, France
e
Hepatology and Liver Intensive Care, INSERM U1149, Hospital Beaujon, Clichy, France
f
Chirurgie Digestive, Endocrinienne et Transplantation Hpatique, Hpital Trousseau, Tours, France
g
AP-HP Hpital Paul-Brousse Centre Hpato-Biliaire and INSERM, Unit 1193, Universit Paris-Sud, UMR S1193, Villejuif, France

a b s t r a c t

The mammalian target of rapamycin (mTOR) inhibitor everolimus is approved for rejection prophylaxis after liver
transplantation. The current article pools the experience of French liver transplant surgeons and physicians in use
of everolimus and, particularly, practical guidance on dosing, appropriate concomitant immunosuppression and
management of adverse events. In terms of indication, introduction of everolimus from week 4 after liver
transplantation, with or without concomitant calcineurin inhibitor (CNI) therapy, offers a signicant renal
benet without loss of immunosuppressive efcacy. De novo treatment with everolimus, either selectively or
systematically, may play a role in the prevention and treatment of recurrence of hepatocellular cancer and
de novo malignancies. For maintenance patients, the most frequent indications for introducing everolimus are in
response to renal dysfunction, recurrent hepatocellular cancer, diabetes, hypertension, or neurotoxicity, or as a
preventative approach to avoid malignancies. Of these, the strongest evidence exists for a renoprotective effect.
However, the low rate of acute rejection following switch of maintenance patients from CNI-based
to everolimus-based therapy means that this can be considered even where robust data are not yet available.
Most adverse events associated with mTOR inhibitors can usually be managed successfully, often with
concentration-controlled dose reductions. Dosing algorithms are provided, with suggestions for target ranges in
specic settings, and treatment strategies for the most common side effects are proposed. Although further
research is required, everolimus has become an established part of the immunosuppressive arsenal for liver
transplant recipients over the last decade. Sharing experience from units which have embraced its use may help
other centers develop their own protocols.
2016 Elsevier Inc. All rights reserved.

1. Introduction de novo and maintenance patients after all types of solid organ
transplantation [5]. Initially, the main emphasis for investigation was
The mammalian target of rapamycin (mTOR) inhibitors, everolimus in kidney [5,6] and heart [7,8] transplantation, but more recently
(EVR) and sirolimus, inhibit intracellular pathways that prevent B- and randomized trials [913] and investigator-led single-center reports
T-cell proliferation [1]. EVR (Certican; Novartis Pharma AG, Basel, have substantially increased the evidence base in liver transplant
Switzerland) has recently been approved in the EU and the USA for pro- recipients [14]. To date, however, this experience has not prompted
phylaxis of organ rejection in patients receiving a liver transplant, in publication of practical guidance on how EVR can be optimally
combination with tacrolimus and steroids [2]. Sirolimus is not currently employed in the setting of liver transplantation.
licensed for use in liver transplantation. Both agents share a similar This article summarizes the expert opinion of French liver transplant
structure, other than addition of an extra hydroxyethyl group in the surgeons and physicians who have been treating patients with EVR for
EVR molecule which gives rise to a shorter half-life [3] and more rapid more than 10 years. Based on published data and personal experience,
attainment of stable blood concentrations [4] compared to sirolimus. it considers the appropriate selection of liver transplant patients for
An extensive series of clinical trials has explored the use of EVR EVR therapy and the practicalities of its use.
within various different immunosuppressive regimens for both
2. Methods
Corresponding author at: AP-HP Hpital Paul-Brousse Centre Hpato-Biliaire, Villejuif,
F-94800, France. Tel.: +33 1 45 59 64 12; fax: +33 145 59 38 57. The authors met on three occasions to develop the manuscript:
E-mail address: faouzi.saliba@pbr.aphp.fr (F. Saliba). (1) to identify topics for inclusion which were each then allocated to a

http://dx.doi.org/10.1016/j.trre.2015.12.003
0955-470X/ 2016 Elsevier Inc. All rights reserved.

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003
2 J. Dumortier et al. / Transplantation Reviews xxx (2016) xxxxxx

single author to undertake a literature research and develop draft text The Modication of Diet in Renal Disease (MDRD) and the Chronic
(2) to present and discuss draft outputs (3) to ne-tune the nal con- Kidney Disease Epidemiology Collaboration (CKD-EPI) formulae
tent, which was agreed by all authors. Each author conducted a litera- are most often used to estimate GFR measurement reliably.
ture review using some or all of the following key words, as 24-h proteinuria or urinary protein/creatinine ratio should be measured
appropriate: mTOR inhibitors, liver transplantation combined with: prior to introduction of EVR. (The H2304 study excluded patents with
pharmacokinetics, EVR, sirolimus, tacrolimus, renal function, hepatocel- urinary protein excretion 0.8 g/day at baseline, and all randomized
lular carcinoma (HCC), hepatitis C, liver brosis, de novo cancer, neuro- trials of early switch to EVR stipulated minimum levels of baseline
toxicity, dyslipidemia, surgery, pregnancy, fertility. Additional renal function, so outcomes are undocumented in these patients.)
references were identied from key review papers. In patients with moderate to severe renal impairment, and those
with proteinuria N0.8 g/day, consider consultation with a nephrologist
3. EVR in the de novo setting and perform renal biopsies when necessary.
Introduction of EVR should take place as early post-transplant as
3.1. Preservation of renal function possible. CNI withdrawal, if undertaken, should be carried out
gradually (e.g. over 8 weeks) and induction therapy may be helpful
Impaired renal function is a well-established complication in pa- to maintain immunosuppressive efcacy during early switch from
tients with end-stage liver disease, and frequently progresses after CNI treatment.
transplantation. A population-based cohort study estimated that 18.1% Induction is also likely to be useful if CNI initiation is delayed, or if
of liver transplant recipients develop chronic renal failure (dened as CNI levels are decreased rapidly, in EVR-treated patients.
estimated GFR [eGFR] b 30 mL/min/1.73m 2) by ve years post-
transplant, and with an associated four-fold increase in mortality [15]. 3.2. Prevention of de novo or recurrent non-HCC malignancies
The causes of post-transplant chronic renal insufciency are multifacto-
rial, but long-term exposure to calcineurin inhibitor (CNI) therapy is an Post-transplant malignancy has become one of the main causes of
important contributor. Renal-sparing strategies have included use of death after liver transplantation, irrespective of the primary indication
monoclonal anti-CD25 antibody induction to delay CNI initiation for transplant [24,25]. The overall standardized incidence ratio for risk
[16,17], addition of mycophenolate mofetil (MMF) [18,19] and, more re- of malignancy compared to the general population has been reported
cently, introduction of mTOR inhibitor therapy to reduce CNI exposure to range from 2.1 to 3.2 [2628] and is signicantly higher when pa-
[9,10] or discontinue CNI entirely either early (b3 months post- tients are transplanted for alcoholic liver disease [29]. The mTOR inhib-
transplant) [11,13] or later [12]. EVR acts synergistically with CNIs itor class can exert an antitumoral effect depending on the type of
[20], allowing CNI exposure to be substantially reduced while maintain- cancer (especially its hypervascularized characteristics). At higher
ing sufcient immunosuppressive activity [10,11,20]. doses, mTOR inhibitors can exert a direct antiproliferative effect on can-
In order to minimize CNI-related nephrotoxicity and preserve long- cer cells, while the lower doses used in transplant recipients are associ-
term renal function, CNI exposure should be tapered as soon as possible ated with an antiangiogenic effect [30]. There is evidence for an anti-
after transplantation, but this needs to be balanced with effective im- neoplastic activity at clinically relevant doses (5 mg/day) [21,31]. In-
munosuppression. In the two-year, multicenter, open-label, random- deed, EVR is indicated for the treatment of renal cell carcinoma, breast
ized H2304 study, patients received standard-exposure tacrolimus cancer, neuroendocrine tumors and angiomyolipoma [3237]. Studies
with steroids for the rst month [9]. At week 4 post-transplant, patients in kidney transplantation have reported a lower incidence of de novo
were randomized to (1) standard-exposure tacrolimus as the control post-transplant malignancies under mTOR inhibitor therapy [3840].
regimen (n = 243), (2) EVR with reduced-exposure tacrolimus (n = Data are more limited in liver transplant populations. One large retro-
245), and (3) EVR with tacrolimus elimination (n = 231). One-year spective single-center analysis of liver transplants performed during
rates of biopsy-proven acute rejection (BPAR) were 10.7% in the control 1996 to 2013 compared the incidence of new-onset post-transplant ma-
group versus 4.1% for EVR with reduced-exposure tacrolimus (p = lignancies in 243 patients who were given EVR for reasons other than
0.005 in favor of the EVR cohort). All cases of BPAR in EVR-treated patients malignancy versus 1182 patients without any mTOR inhibitor treat-
were graded borderline or mild [9]. The between-group difference in the ment [41]. After a median follow-up of 1740 days, the incidence of
rate of BPAR was sustained at two years (13.3% versus 6.1%, p = 0.010) new-onset malignancies was 0.2% in the EVR-treated group and 3.4%
[9]. A recent review by Klintmalm et al. concluded that EVR-based immu- in the patients without an mTOR inhibitor. EVR-free immunosuppres-
nosuppression achieves similar rates of acute rejection, graft and patient sion was found to be an independent predictor for risk of malignancy.
survival to conventional regimens comprising CNI and MMF [14]. Conrmatory studies are lacking, however. Also, since increased CNI ex-
Entirely CNI-free immunosuppression with EVR and mycophenolic posure is associated with higher risk of solid cancers after liver trans-
acid (MPA), however, may not provide adequate protection against rejec- plantation [41,42], it is not clear to what extent any reduction in
tion [21], and even early CNI withdrawal must be undertaken cautiously. malignancies may be due to inhibition of the mTOR pathway, and
In the tacrolimus elimination arm of the H2304 study, randomization was what contribution arises from reduced CNI exposure in the presence
discontinued prematurely due to high rates of BPAR clustered around the of an mTOR inhibitor.
time of tacrolimus withdrawal. In that trial, CNI was withdrawn abruptly For patients receiving a liver transplant due to malignancies such as
after month 4 post-transplant. In contrast, the randomized PROTECT hilar cholangiocarcinoma or liver metastases from neuroendocrine tu-
study found that more gradual CNI elimination over an eight-week period mors, it is unknown whether an mTOR inhibitor-based immunosup-
starting at week 4, coupled with basiliximab induction, found no loss of pressive regimen could inuence the rate of recurrence [43]. The
efcacy after CNI discontinuation [11]. relative rarity of such cases, and the long follow-up required, are bar-
Early introduction of EVR, either with ongoing reduced-exposure riers to reaching a denitive answer.
CNI or CNI withdrawal, has consistently shown a clinically relevant
renal benet versus a standard CNI regimen which appears to be 3.2.1. Practical implications
sustained to at least three years after liver transplantation (Table 1). At present no rm conclusions can be drawn regarding prevention
of de novo or recurrent cancers using EVR therapy and this cannot
3.1.1. Practical implications currently be regarded as a rm indication for EVR after liver
Renal function (eGFR) should be evaluated prior to liver transplant, transplantation.
immediately post-transplant, at months 1, 3, 6 and 12 and subse- Data available from renal transplant registries are highly encouraging;
quently twice a year similar data from liver transplant populations are awaited.

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003
J. Dumortier et al. / Transplantation Reviews xxx (2016) xxxxxx 3

Table 1
Renal outcomes under everolimus (EVR)-based immunosuppression in prospective trials of liver transplant recipients.

Study Reference Follow-up Treatment group N Mean (SD) eGFR, Mean difference for change p Value for
(months) mL/min/1.73m2 in eGFR from (EVR control) difference

Early EVR + reduced CNI


H2304 De Simone 2012 [9] 12 EVR + low TAC 245 80.9 (27.3) 8.5 (97.5% CI 3.7, 13.3) b0.001
TAC Control 243 70.3 (23.1
Saliba 2013 [10] 24 EVR + low TAC 245 74.7 (26.1) 6.7 (97.5% CI 1.9, 11.4) 0.002
TAC Control 243 67.8 (21.0)
Fischer 2015 [22] 36 EVR + low TAC 106a 78.7 (25.7) 8.5 b0.005
TAC Control 125a 63.5 (18.3)

Early EVR + CNI discontinuation


PROTECT Fischer 2012 [11] 12 Switch CNI to EVR by M4 101 80.3 (26.4) 7.8 (95% CI 0.1, 14.7) 0.021
CNI Control 102 72.1 (24.5)
Sterneck 2014 [23] 36 Switch CNI to EVR by M4 41a 77.5 (23.4) 9.4 (95% CI -0.4, 19.8) 0.053
CNI Control 40a 67.9 (21.8)
Single-center prospective Masetti 2010 [13] 12 EVR from day 10, 52 87.7 (26.1)b - -
CNI withdrawn after day 30
CsA control 26 59.9 (12.6)b

Maintenance patients: Conversion from CNI to EVR


RESCUE De Simone 2009 [12] 6 Switch from CNI to EVR 72 52.8 (12.2)c -1.1 (95% CI -3.9, 1.9)c
CNI control 73 52.7 (10.4)c 0.463
12 Switch from CNI to EVR 72 53.8 (12.8)c -
CNI control 73 52.5 (12.7)c

CI, condence interval; CNI, calcineurin inhibitor; eGFR, estimated glomerular ltration rate (Modication of Diet in Renal Disease formula); EVR, everolimus; SD, standard deviation; TAC,
tacrolimus.
a
Study extension population.
b
p b 0.001 for between-group difference in observed eGFR at month 12.
c
Creatinine clearance (Cockcroft-Gault formula).

The exception is neuroendocrine tumors, for which evidence from facilitate CNI reduction or elimination. An increase in rejection with
non-transplant populations suggests that EVR can be an effective the requirement for intensication of immunosuppression would,
treatment option [35]. however, be counterproductive due to the enhanced risk of tumor
recurrence.
3.3. Recurrence of HCC Two options can be considered:

HCC is now a leading indication for liver transplantation but because


post-transplant tumor recurrence is almost invariably fatal within two (i) Detection on explant pathology and prognosis is poor (poor dif-
years, patients are selected according to dened risk factors such as ferentiated tumor, microvascular invasion, outside Milan criteria;
those specied in the well-established Milan criteria [44]. Selection the current trend in France), i.e. the risk of recurrence was under-
criteria are based on the morphological characteristics of the carcinoma stated pre-transplant.
and, to a lesser extent, on biomarker levels (e.g. alpha-fetoprotein) (ii) Systematic introduction of EVR to support CNI minimization/
which reect tumor biological behavior. The criteria are not compre- withdrawal in all cases of liver transplant for HCC, irrespective
hensive and do not take into account the type and intensity of the im- of explant pathology. Such an approach is supported by the US ex-
munosuppressive regimen. There is growing evidence to suggest, for perience in which HCC recurrence was reduced under sirolimus
example, that the risk of HCC recurrence increases with cumulative therapy for patients transplanted within Milan criteria [59].
CNI exposure [45,46] and use of ATG induction [47].
Interest is growing in a role for EVR in preventing post-transplant
HCC recurrence [48]. The mTOR pathway is activated in 3040% For either CNI reduction strategy, start EVR by the end of month 1,
of HCC cases [49] and preclinical data suggest that clinical exposure then when EVR trough level reaches 38 ng/mL reduce CNI dose
levels are adequate to exert antitumor activity in HCC [31], with anti- by half then gradually taper CNI (target trough levels of tacrolimus:
proliferative effects on HCC cell lines sustained even with concomitant 35 ng/mL) for 612 months.
tacrolimus administration [50]. It would be appealing to initiate EVR Consider progressing to EVR monotherapy after year 1 depending on
selectively in cases where there is pathological proof of mTOR pathway the risk/benet ratio for tumor recurrence, and targeting EVR trough
activation, but this remains impractical at present. levels of 610 ng/mL if CNI is withdrawn. EVR exposure below
Clinically, data relating to an effect of mTOR inhibition are largely re- 6 ng/mL may be considered in low-risk maintenance patients receiving
stricted to retrospective and non-randomized prospective analyses [48]. MPA therapy.
These suggest a benet for HCC recurrence rates versus standard CNI Addition of MPA therapy may be helpful to prevent rejection in
therapy [48,5155]. In some cases the patient population was extremely patients switched from CNI to EVR during the rst year post-
selected [51,56,57], but EVR has also been shown to offer advantages for transplant.
HCC recurrence and post-recurrence survival for patients transplanted A few centers have attempted very early introduction of EVR post-
outside the Milan criteria [58]. A randomized controlled trial using transplant but few data are available on the safety of this strategy.
EVR in this setting is ongoing (NCT01888432).
3.4. Hepatitis C virus (HCV) recurrence
3.3.1. Practical implications
Reducing the risk of post-transplant HCC recurrence should include Prospective trials investigating the role of mTOR inhibitors after liver
avoidance of anti-thymocyte globulin induction, with early CNI mini- transplant for HCV-related disease are lacking, and the available evi-
mization or even withdrawal. mTOR inhibition can be helpful to dence is inconclusive. However, an analysis from the HCV-positive

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003
4 J. Dumortier et al. / Transplantation Reviews xxx (2016) xxxxxx

subpopulations in three controlled studies showed a trend to less Other, non-comparative studies have described a signicant improve-
brosis progression in patients receiving EVR with reduced CNI versus ment in renal function following switch from CNI to EVR which appears
standard immunosuppression [60]. A non-randomized study cohort in to be sustained to at least one year after conversion [65,72]. Overall, the
de novo liver transplant patients reported reduced HCV replication in available data indicate that maintenance patients can be switched from
sirolimus-treated patients with previous HCV infection [61], but no CNI to EVR in response to renal deterioration without an increase in
effect on viral load was observed in the H2304 study [9]. Three rejection risk [12,65,72] but that late CNI withdrawal is less effective
published trials of mTOR inhibition in de novo patients have observed in ameliorating loss of renal function [73].
delayed brosis in HCV-positive recipients [6163].
However, introduction of new direct-acting antiviral (DAA) therapies such 4.1.1. Practical implications
as sofosbuvir, simeprevir, sofosbuvir + daclatasvir, sofosbuvir + ledipasvir In maintenance liver transplant patients who have developed CNI-
and ombitasvir/paritaprevir/ritonavir + dasabuvir has largely obviated related renal impairment, introduce EVR and taper CNI exposure.
any inuence of the immunosuppressive regimen. Of note, unlike Withdraw CNI in patients who are more than 1 year post-transplant
sofosbuvir which is mainly metabolized by the kidney, the coadminis- unless there is a high risk of rejection (e.g. previous rejection
tration of these drugs with EVR has not been studied but as with CNIs, episodes, autoimmune disease).
they may increase EVR concentrations either due to CYP3A4 and/or P- Maintain EVR trough levels toward the upper end of a 38 ng/mL
glycoprotein inhibition [64]. If used, therapeutic drug monitoring target range if CNI is discontinued, and consider introduction of
of EVR should be performed when DAA therapy is started and stopped, MMF to maintain immunosuppressive efcacy in the absence of CNI.
together with careful monitoring of DAA-related side effects
and toxicities.
4.2. CNI-induced neurotoxicity
3.4.1. Practical implications
EVR-based immunosuppression is an appropriate option in HCV- Post-operative neurological complications such as confusion, sei-
positive liver transplant patients and may delay brosis progression, zures and tremor occur in approximately 1015% of liver transplant pa-
but any such an effect is less relevant if DAA therapy is routinely tients [74,75], with rare (b1%) severe neurological complications such
administered to HCV-positive recipients. as cerebral hemorrhage, ischemic stroke, and posterior reversible
A summary of drugdrug interactions between DAA products encephalopathy syndrome (PRES) [75,76]. CNI therapy is known to be
and immunosuppressive agents (including everolimus) is shown associated with neurotoxic adverse events such as tremor, confusion
in Table 2. and seizures, which usually resolve or improve signicantly with dose
reduction. Switch from CNI to mTOR inhibitor therapy is undertaken
4. EVR in the maintenance setting due to neurotoxicity in approximately 15% of cases [77]. CNI agents
are also the principal cause of PRES, and most cases improve if the
The literature contains reports of EVR introduction at various time patient is converted to an mTOR inhibitor at an early stage [77].
points after liver transplantation, for a range of indications [6571].
The authors' most frequent reasons for starting EVR in maintenance 4.2.1. Practical implications
patients are in response to renal dysfunction, recurrent HCC, diabetes, CNI-induced PRES mandates emergency discontinuation of CNI
hypertension, or neurotoxicity, or for the prevention or management therapy. Switching to an mTOR inhibitor is generally considered
of malignancies. In the absence of data, the authors do not recommend the best option to prevent further progression while providing
EVR to treat either acute or chronic rejection. effective immunosuppression.
One prospective study [12] and retrospective series [65,6971] have
reported a low acute rejection rate after switch from CNI, as might be
4.3. Management of post-transplant malignancy
expected in maintenance patients. Rates of adverse events and discon-
tinuations were acceptable.
4.3.1. HCC recurrence
Management of HCC recurrence is based on the premise that
4.1. Patients with deteriorating renal function
treatment of an advanced oncological disease with an extremely poor
prognosis is a greater clinical priority than prevention of rejection.
In a 12-month trial of 145 liver transplant patients with CNI-
Three options can be considered, irrespective of the surgical possibilities
related renal impairment (baseline creatinine clearance in the range
for tumor resection:
2060 mL/min), De Simone et al. found no renal benet in the group
randomized to EVR with CNI discontinuation (80%) or reduction (20%) (i) Introduce EVR with immediate CNI withdrawal.
versus a CNI-continuation control arm [12]. Results were complicated (ii) Introduce EVR if mTOR pathway activation is detected on
by the fact that 77% of controls also received CNI dose reductions; addi- biopsies of tumor tissue. However, this approach is not routinely
tionally, the mean time post-transplant was more than three years. available.

Table 2
Drugdrug interactions between direct-acting antiviral (DAA) therapies against HCV infection and immunosuppressive agents [64]. More details are available at www.hep-
druginteractions.org [64].

Simeprevir Daclatasvir Sofosbuvir Sofosbuvir + ledipasvir 3D (ritonavir-boosted paritaprevir + ombitasvir + dasabuvir)

Azathioprine
Cyclosporine +++ + +
Everolimus + + + +++
Mycophenolic acid +
Sirolimus + + +
Tacrolimus + + +

, No clinically signicant interaction expected.


+, Potential interaction which may require a dosage adjustment, altered timing of administration or additional monitoring.
+++, These drugs should not be co-administered.

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003
J. Dumortier et al. / Transplantation Reviews xxx (2016) xxxxxx 5

(iii) Combine EVR with sorafenib on the grounds that pre-clinical 5. Management of EVR-related adverse events
studies and case reports have suggested a synergistic effect
between the two therapies [78,79], although a cautious approach Class-related adverse events and clinical complications associated
is required due to safety concerns. with mTOR inhibitors are well documented in the literature [68] and
can usually be managed successfully [87]. These include inhibition of
4.3.1.1. Practical implications. wound healing, peripheral edema, mouth ulceration, dermatological
Start EVR at 1 mg b.i.d., targeting trough levels of 812 ng/mL side effects such as acne, hypercholesterolemia, proteinuria and, rarely,
according to patient tolerability, and discontinue CNI as soon as possible. interstitial pneumonitis. Some adverse effects are less frequent and less
If the patient is receiving MMF, assess if this should be continued severe if appropriate starting doses are used and if therapeutic blood
based on local practices. levels are carefully controlled [68]. Many side effects are dose depen-
Consider combination therapy with sorafenib, although poor dent and frequently the rst response is to check EVR exposure and con-
tolerability of sorafenib when combined with EVR may necessitate sider a dose reduction. Suggestions for management are outlined in
a sorafenib starting dose of 200 mg b.i.d. adjusted according to Table 3, and specic adverse events are discussed below.
therapeutic drug monitoring.
5.1. Wound healing complications
4.3.2. other solid tumors
Small studies have examined switching liver transplant recipients In a rat model, EVR has been shown to signicantly inhibit the cell
with de novo solid tumors from a CNI to sirolimus or EVR [69,8082]. proliferation and new collagen deposition that is required for normal
Converting to sirolimus with rituximab was associated with improved wound repair [95]. Clinically, the effect appears to be dose dependent.
long-term disease-free survival after treatment of lymphoproliferative In a pooled analysis of three prospective studies in kidney transplanta-
disorders in one report [83]. In a small retrospective study, Gomez- tion, wound healing complications were more frequent when EVR
Camarero et al. showed that patients presenting de novo malignancies trough levels of 610 ng/mL were targeted versus a MMF-treated con-
after liver transplantation survived longer when switched to EVR [81]. trol group, but comparable to controls when the target trough level
In a single-center study of 83 patients transplanted for alcoholic liver was 38 ng/mL [96].
disease, 38 patients started EVR therapy with CNI discontinuation in Surgically, liver transplantation is more complex than kidney en-
the majority of cases (64.1%) [69]. One- and ve-year survival was graftment, with more frequent complications. Wound healing events
77.4% and 35.2% in the EVR cohort compared to 47.2% and 19.4% in the after liver transplantation can include thrombosis of the hepatic artery
control group (p = 0.003) [69]. or portal vein, incisional hernias, biliary stulas, or delayed cutaneous
Converting from CNIs to mTOR inhibitors is part of the routine wound repair.
treatment of post-liver transplant Kaposi sarcoma [84]. In the H2304 study, introduction of EVR with reduced tacrolimus at
There are no clinical studies suggesting a signicant protective effect four weeks post-transplant was associated with a similar 12-month rate
of mTOR inhibition against recurrence of pre-existing malignancies of wound healing complications to the tacrolimus control arm (11.0%
after liver transplantation. Nevertheless, EVR may be considered a versus 8.3%) based on a target EVR range of 38 ng/mL [9]. Randomized
therapeutic option for some malignant tumors such as kidney and trial data relating to EVR therapy from the day of transplant are not
breast cancers, because EVR is approved in these settings. available. There are published cases of liver transplant patients under-
going major abdominal or thoracic surgical procedures while receiving
4.3.2.1. Practical implications. an unchanged dose of an mTOR inhibitor [97] but this has not been ex-
Conversion to EVR (target 510 ng/mL) could be considered in amined rigorously.
patients with a poor metastatic prognosis, to increase short-term
survival. Concomitant MMF is an option according to local practice, 5.1.1. Practical implications
but is contraindicated in patients with severe chemotherapy- Delay EVR introduction until complete surgical healing if the patient
induced anemia. has surgical complications, infections or a general condition that
CNI should be tapered and discontinued within a few months of may negatively affect wound healing, such as prolonged hyperventi-
starting EVR. CNI discontinuation can be considered depending on lation post-surgery.
the severity and prognosis of the cancer and the risk of rejection. In rare cases, particularly with T-tube external biliary drainage,
The authors have anecdotal occasional negative experiences com- consider delaying EVR until months 3 and 4.
bining EVR with certain chemotherapies (e.g. gemcitabine combined It may be advisable to avoid early EVR in malnourished patients with
with oxaliplatin, bevacizumab) low body mass index (BMI) (e.g. b 20 kg/m2) and in obese patients
Caution should be taken in EVR-treated patients given radiation (BMI N 35 kg/m 2). Ensure correct wound healing (including signs
therapy because of the enhanced risk of tissue burning or injury. of wound budding) before starting EVR in these patients, since low
or high BMI can increase risk of healing complications.
4.3.3. Skin cancer Consider interrupting EVR 45 days before major elective surgery
Ducroux et al. recently reported a 13.5% prevalence of skin cancers in with a compensatory increase of CNI dosing (to allow for EVR clear-
371 liver transplant patients over a median follow-up of 8.2 years [85], ance based on a half-life of 28 h), reintroducing EVR approximately
comparable to that observed after kidney transplantation. A random- one month after the procedure.
ized trial in kidney transplant patients has shown that switch from
CNI therapy to sirolimus signicantly reduced the risk of cutaneous 5.2. Dyslipidemia
squamous cell carcinoma recurrence [86]. Similar data are not available
for a liver transplant population. Hyperlipidemia is common in organ transplant recipients, and is ex-
acerbated by CNI agents and mTOR inhibitors [91,98]. The dyslipidemic
4.3.3.1. Practical implications. inuence of mTOR inhibitor therapy has diminished using contempo-
Switch from CNI to an mTOR inhibitor in liver transplant patients rary dosing protocols which avoid a loading dose and deliver lower,
with previous squamous cell carcinoma, and possibly other forms concentration-controlled exposure compared to early trials, but an ef-
of non-melanoma skin cancer, is of potential interest. fect persists [91]. Liver transplant recipients, unlike heart transplant pa-
More data are required in the liver transplant population and for tients, do not routinely receive statins and total cholesterol levels are
other types of skin cancer. frequently toward the upper end of normal. Most lipid-related adverse

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003
6 J. Dumortier et al. / Transplantation Reviews xxx (2016) xxxxxx

Table 3
Incidence and management of everolimus (EVR)-related adverse events.

Adverse event Incidence in liver transplantation Management

Wound healing complications 2.215% (may be less prevalent with EVR vs. sirolimus) Withdraw EVR 45 days before major or parietal surgery and reintroduce
after 30 days (or after complete healing) [88]
Peripheral edema 717.6% [9,13,65,68,89] Consider the possible contribution of concomitant drugs
Evaluate response to diuretics
Consider EVR reduction (particularly if trough concentration is high)
or EVR withdrawal if serious periorbital or angioedema
Mouth ulcers Up to 20% of de novo patients [68] Warn patients of this side effect
19% in maintenance patients [65] Rule out viral/fungal infection
Topical analgesic
Topical steroids
Betamethasone can be prescribed as a preventive treatment,
to be used as soon as mucosal lesions appear
Dermatological disorders Acne, rash and eczema reported in 1420% [9,65,68] Consider EVR dose reduction
Rule out infection
May resolve spontaneously within a few weeks
Topical treatment e.g. emollients
Consider reintroducing low-dose steroids [90]
Refer to a dermatologist if required
Hematological disorders Anemia (13%), leukopenia (9%), Consider EVR reduction (particularly if trough concentration is high)
thrombocytopenia (6%) or EVR withdrawal
(may be more frequent under sirolimus) [65,87] Rule out other causes (mycophenolic acid, ganciclovir/valganciclovir or other drugs,
vitamin or iron deciency, viral infections)
Evaluate reduction or withdrawal of MMF
Erythropoiesis stimulation/iron therapy
GM-CSF treatment
Thrombopoietin receptor agonists
Hyperlipidemia 743% [14,91] Suggest lifestyle changes
Initiate lipid-lowering therapy with statins and/or omega-3 fatty acids
Proteinuria 5.518% in maintenance patients Do not switch to mTOR inhibitor therapy if 24 h proteinuria is 0.8 g/L [50]
converted to EVR [65,68] Perform renal biopsy if necessary [92]
Consider reduction or withdrawal of EVR if possible
Initiate treatment with an angiotensin-converting enzyme inhibitor or
angiotensin II receptor blocker [93]
Interstitial pneumonitis 0.40.5% of solid organ tx patients overall [94] Evaluate reduction of EVR exposure or withdrawal if symptoms persist
Initiate steroid treatment if infection and cancer are ruled out
Refer to lung specialist
Consider thoracic scan as a reference examination

CNI, calcineurin inhibitor; EVR, everolimus; GM-CSF, granulocyte-macrophage colony-stimulating factor; MMF, mycophenolate mofetil; mTOR, mammalian target of rapamycin; tx,
transplantation.

events under EVR are mild and can be managed successfully [87], and 6. Dosing and monitoring of EVR
are not generally regarded as a barrier to EVR therapy.
6.1. De novo liver transplant patients (Fig. 1a)
5.2.1. Practical implications
6.1.1. Initiation of EVR and target trough concentration
Monitor lipid prole and manage dyslipidemia closely with combi-
Liver transplantation is associated with a relatively high
nations of diet modication, appropriate statin therapy, ezetimibe,
rate of post-transplant healing complications and surgical re-
sh oils, brates or extended release niacin and physical activity.
interventions, such as laparotomy. The product license therefore
recommends that EVR be initiated approximately four weeks after
liver transplantation at a starting dose of 1.0 mg twice daily, targeting
5.3. Pregnancy and male fertility
a trough concentration 38 ng/mL, in combination with tacrolimus
and steroids.
Retrospective studies, mostly from the early high-dose era of
In practice, the authors usually start EVR at either 0.75 mg or
sirolimus therapy, have suggested a potential negative effect of mTOR
1.0 mg twice daily, increasing the dose progressively as required,
inhibitors on male gonadal function [99]. If male patients with a low
usually to a maximum of 2 mg b.i.d. The ideal target trough level
sperm count under sirolimus wish to reproduce, they may be successful
should be 56 ng/mL, particularly during the early post-
under sirolimus or can be switched to another drug for a period. Tissue
transplant period (i.e. the rst four months) but certainly levels
banks may not accept sperm from donors with viral hepatitis. The role
should not fall below 3 ng/mL. Dose adjustments are required ac-
of mTOR inhibitors in pregnancy is poorly documented, being limited
cording to trough level, individual response, tolerability, and
to single case reports that have not described signicant fetal effects
changes in co-medications [68] but should be made only after measur-
[100]. The authors are cautiously optimistic about breast-feeding
ing trough levels 45 days after reaching steady state. Trough level
under mTOR inhibitor therapy, but more data are required. Given the
should be monitored after any dose change and then at regular consul-
small number of cases and the variety of immunosuppressive regimens,
tation visits.
only broad-based registry participation will provide the evidence need-
In patients with renal impairment at time of transplant, inter-
ed to support more condent recommendations.
leukin 2 (IL-2) receptor antagonist induction therapy with
basiliximab should be followed by triple therapy comprising
5.3.1. Practical implications MPA, low-dose steroids and tacrolimus delayed until days 35 (targeting
EVR should be stopped if pregnancy is planned (at least six weeks 610 ng/mL during the rst month). EVR can then be initiated from
before conception) or occurs. week 4.

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003
J. Dumortier et al. / Transplantation Reviews xxx (2016) xxxxxx 7

A Liver Tx

Renal impairment
TAC MPA + steroids CsA MPA + steroids
at time of transplant

Basiliximab induction
Delayed CNI (day 3-5)
MPA + low-dose steroids

EVR 1.0mg bid EVR <1.0mg bid


Week 4 Discontinue MPA overnight Discontinue MPA overnight
Continue steroids Continue steroids

Measure EVR trough Measure EVR trough


level after 4-5 days level after 4-5 days

Measure EVR trough When EVR trough is When EVR trough is


level 4-5 days after 3-8ng/mL, progressively 3-8ng/mL, rapidly reduce
any dose change reduce TAC to 3-5ng/mL CsA to 50-100ng/mL

Target EVR trough Target EVR trough


Month 3 level 5-6ng/mL* level 5-6ng/mL*
(minimum 3ng/mL**) (minimum 3ng/mL**)

* Particularly to month 4 post-transplant


** Increase dose if <3ng/mL and measure trough level 4-5 days later

B Maintenance patients
(>6 months post-transplant)

Reduced CNI EVR monotherapy


(only >1 year post-transplant)

CNI MPA + steroids CNI MPA + steroids

EVR 0.75 bid EVR 1.0 mg bid


Day 0 Discontinue MPA overnight Discontinue MPA overnight
Continue steroids Continue steroids

Increase dose if EVR Measure EVR trough


Day 7 trough level <3ng/mL level after 4-5 days

Measure EVR trough When EVR trough is


level every 4-5 days to When EVR trough is
3-8ng/mL, progressively 6-12ng/mL, taper &
day 21 and after reduce CNI (e.g. discontinue CNI
any dose change TAC 3-5ng/mL)

Target EVR Target EVR trough


trough level >3ng/mL* level 6-12ng/mL**

* Increase dose if <3ng/mL and measure trough level 4-5 days later
** Increase dose if <6ng/mL and measure trough level 4-5 days later

Fig. 1. Suggested approach to everolimus (EVR) dosing in (a) de novo and (b) maintenance liver transplant patients.

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003
8 J. Dumortier et al. / Transplantation Reviews xxx (2016) xxxxxx

Table 4
Areas for future research relating to everolimus therapy in liver transplantation.

Topic Rationale Study data required in Ongoing study/studies


liver transplantation

Earlier introduction of EVR Randomized trials to date have not Prospective, randomized data HEPHAISTOS
(before week 4 post-transplant) started CNI reduction/withdrawal b30 days (NCT01551212) [103]
Prevention of HCC recurrence Limited data to date suggest benet [48] Prospective data H2307 (everolimus)
and survival after tx for HCC e.g. casecontrolled (NCT01888432)
cohort studies SILVER study (sirolimus)
(NCT003558662)
Prevention of malignancy/survival Only retrospective data available to Prospective data from larger SIMCER study (EVR)
benet for de novo post-tx cancer date in liver transplantation [69] studies e.g. cohort or (NCT01625377)
case-control studies extension phase
(CERTITUDE)
Prevention of skin cancer Switch from CNI to mTOR inhibitor Prospective data -
recurrence after liver tx inhibits SCC recurrence after kidney tx [86] e.g. case-controlled cohort studies
De novo DSA Switch from CNI to EVR with low-dose Prospective data evaluating
MPA steroids at M3 associated with rate of de novo DSA under
under-immunosuppression and increased EVR + MPA (CNI-free) vs. EVR
DSA in kidney transplantation [104] with reduced CNI
Dyslipidemia EVR therapy is associated with increased Long-term data on SIMCER study (EVR)
lipid levels but antiproliferative effects may be cardioprotective cardiovascular event rate (NCT01625377)
extension phase (CERTITUDE)

CNI, calcineurin inhibitor; DSA, donor-specic antibodies; EVR, everolimus; HCC, hepatocellular carcinoma; MPA, mycophenolic acid; mTOR, mammalian target of rapamycin; SCC, squamous
cell carcinoma; tx, transplantation.

6.1.2. Switch from MPA dose of between 0.25 and 0.75 mg b.i.d., slowly increasing the dose over
MPA can be safely discontinued abruptly (overnight) when EVR approximately three weeks to achieve the same targets and continuing
is introduced with concomitant CNI, as demonstrated in the H2304 monitoring at routine consultation visits.
trial [9]. For patients in whom EVR is introduced in response to renal deteri-
oration, the RESCUE study showed a signicant inverse relationship be-
6.1.3. Concomitant MPA therapy tween baseline renal function and the renal benet from switch to EVR
Randomized trials to date have not included MPA therapy in [12], so EVR should be considered as early as possible.
everolimus-based CNI-free treatment regimens [1113]. The ongoing
SIMCER study, however, randomizes de novo liver transplant patients 6.2.2. Switch from MPA
at one month post-transplant to either switch from tacrolimus to everoli- As in the de novo setting, MPA can be stopped abruptly when EVR is
mus while continuing MPA therapy (steroids) or to remain on tacrolimus introduced in maintenance patients.
with MPA (steroids), all with basiliximab induction (NCT01625377). The
primary endpoint is change in eGFR from randomization to month 6 6.2.3. EVR monotherapy
post-transplant, and results are awaited with interest. The authors consider that EVR monotherapy is an option after the
rst post-transplant year in selected patients, with very low rates of re-
6.1.4. Concomitant CNI therapy jection reported [12,65]. In the absence of concomitant CNI or MPA, the
CNI dose should be progressively reduced only after EVR trough target EVR range should be 610 ng/mL.
level reaches the range 38 ng/mL. The nal tacrolimus target range is
35 ng/mL [9]. EVR exposure is increased in patients receiving concom- 7. Future prospects
itant cyclosporine A (CsA) compared to tacrolimus [101,102]. In CsA-
treated patients, the authors start EVR at a dose less than 1 mg/day Certain questions remain unanswered about EVR therapy in liver
twice daily and rapidly reduce the CsA dose (targeting 50100 ng/mL) transplantation and novel indications for its use that require explora-
after EVR trough level reaches 38 ng/mL. tion. Several clinical studies are underway which may shed light on
these questions (Table 4).
6.1.5. EVR monotherapy There is a particular need for further research into the potential ef-
EVR monotherapy is not generally recommended during the rst fects of EVR therapy on recurrence of HCC, and recurrence or de novo de-
12 months after liver transplantation, due to an increased rejection velopment of skin and non-skin cancers after liver transplantation
risk [9], although earlier monotherapy may be appropriate in certain potentially one of the most interesting aspects of EVR-based immuno-
cases e.g. onset of malignancy. CNI withdrawal, if undertaken, should suppression. Experience from kidney transplantation cannot necessarily
be carried out gradually (e.g. over eight weeks) and induction therapy be extrapolated.
may be helpful to maintain immunosuppressive efcacy, as in the Certain safety aspects, notably the risk of de novo donor-specic
PROTECT study protocol [11]. antibody (DSA) production after CNI withdrawal, and the balance
of conventional cardiovascular risk factors versus potentially
6.2. Maintenance liver transplant patients (Fig. 1b) cardioprotective effects associated with mTOR inhibition [88], also
merit additional investigation. Further studies and clinical experience
6.2.1. EVR initiation and target trough concentration may also help to improve rates of EVR discontinuation, as familiarity
In the RESCUE trial, EVR was started in patients who had undergone with adverse events and their management grows [68], and ideally
liver transplantation at least 12 months previously at an initial dose of may help to identify patients at risk for events such as proteinuria in
0.75 mg b.i.d., adjusted after two weeks to target a trough concentration order to target EVR use more accurately.
of 38 ng/mL in CNI-treated patients and 612 ng/mL if CNI therapy was Lastly, there is growing interest in dening possible biomarkers
eliminated [12]. To improve tolerance, the authors usually start EVR at a which could identify patients in whom EVR is likely to be most

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003
J. Dumortier et al. / Transplantation Reviews xxx (2016) xxxxxx 9

benecial, for example regarding prevention of HCC recurrence [105] [12] De Simone P, Metselaar HJ, Fischer L, et al. Conversion from a calcineurin inhibitor
to everolimus therapy in maintenance liver transplant recipients: a prospective,
and minimizing intolerance, although this is still at a very early stage. randomized, multicenter trial. Liver Transpl 2009;15:12629.
Although further research is required, EVR has become an [13] Masetti M, Montalti R, Rompianesi G, et al. Early withdrawal of calcineurin inhibi-
established part of the immunosuppressive arsenal for liver transplant tors and everolimus monotherapy in de novo liver transplant recipients preserves
renal function. Am J Transplant 2010;10:225262.
recipients over the last decade. Centers which have embraced its use [14] Klintmalm GB, Nashan B. The role of mTOR inhibitors in liver transplantation:
have learned strategies for EVR initiation and dosing, and optimal con- reviewing the evidence. J Transplant 2014;2014:845438.
comitant regimens, while managing side effects to minimize intolerance. [15] Ojo AO, Held PJ, Port FK, et al. Chronic renal failure after transplantation of a
nonrenal organ. N Engl J Med 2003;349:93140.
The experience summarized here may help other centers develop their [16] Neuberger JM, Mamelok RD, Neuhaus P, et al. Delayed introduction of reduced-
own strategies for incorporating EVR into local liver transplant care. dose tacrolimus, and renal function in liver transplantation: the ReSpECT study.
Am J Transplant 2009;9:32736.
[17] Calmus Y, Kamar N, Gugenheim J, et al. Assessing renal function with daclizumab
Conict of Interest induction and delayed tacrolimus introduction in liver transplant recipients. Trans-
plantation 2010;89:150410.
[18] Boudjema K, Camus C, Saliba F, et al. Reduced-dose tacrolimus with mycophenolate
The authors have the following conicts of interest to declare in mofetil vs. standard-dose tacrolimus in liver transplantation: a randomized study.
relation to Novartis in the last three years: Am J Transplant 2011;11:96576.
Jerme Dumortier has received a grant, research support and [19] Karie-Guigues S, Janus N, Saliba F, et al. Long-term renal function in liver transplant
recipients and impact of immunosuppressive regimens (calcineurin inhibitors
speaker's fees from Novartis and is a member of a Novartis advisory alone or in combination with mycophenolate mofetil): the TRY study. Liver Transpl
committee. 2009;15:108391.
Sebastian Dharancy has received a grant and speaker's fees from [20] Schuurman HJ, Cottens S, Fuchs S, et al. SDZ RAD, a new rapamycin derivative: syn-
ergism with cyclosporine. Transplantation 1997;64:325.
Novartis, Gilead, BMS and is a member of advisory committees for
[21] Jimnez-Prez M, Gonzlez Grande R, Rando Muoz FJ, et al. Everolimus plus my-
Novartis, Chiesi and Gambro advisory committee. cophenolate mofetil as initial immunosuppression in liver transplantation. Trans-
Yvon Calmus has participated in advisory boards for Novartis and plant Proc 2015;47:902.
[22] Fischer L, Saliba F, Kaiser GM, et al. Three-year outcomes in de novo liver transplant
been an investigator for clinical studies funded by Novartis.
patients receiving everolimus with reduced tacrolimus: follow-up results from a
Christophe Duvoux has participated in advisory boards for Novartis. randomized, multicenter study. Transplantation 2015;99:145562.
Franois Durand has been a consultant for Novartis, Astellas, Gilead [23] Sterneck M, Kaiser GM, Heyne N, et al. Everolimus and early calcineurin inhibitor
and BMS and has received grants from Astellas and Gilead. withdrawal: 3-year results from a randomized trial in liver transplantation. Am J
Transplant 2014;14:70110.
Ephrem Salame has participated at advisory boards for Astellas, [24] Adam R, Karam V, Delvart V, et al. Evolution of indications and results of liver trans-
Novartis, Chiesi, Gilead, Sano and Roche. plantation in Europe. A report from the European Liver Transplant Registry (ELTR).
Faouzi Saliba has received speaker fees and/or research funding J Hepatol 2012;57:67588.
[25] Watt KD, Pedersen RA, Kremers WK, Heimbach JK, Charlton MR. Evolution of
from, and/or has participated at advisory boards for, Novartis, Astellas, causes and risk factors for mortality post-liver transplant: results of the NIDDK
Roche, Genzyme, Merck Sharp & Dohme, Gilead, Janssen, Gambro and long-term follow-up study. Am J Transplant 2010;10:14207.
Vital Therapies. [26] Aberg F, Pukkala E, Hckerstedt K, Sankila R, Isoniemi H. Risk of malignant neo-
plasms after liver transplantation: a population-based study. Liver Transpl 2008;
14:142836.
Acknowledgements [27] Herrero JI, Lorenzo M, Quiroga J, et al. De novo neoplasia after liver transplantation:
an analysis of risk factors and inuence on survival. Liver Transpl 2005;11:8997.
[28] Engels EA, Pfeiffer RM, Fraumeni Jr JF, et al. Spectrum of cancer risk among US solid
The authors would like to thank Pr Georges-Philippe Pageaux of organ transplant recipients. JAMA 2011;306:1891901.
Saint Eloi University Hospital, University of Montpellier I, Montpellier, [29] Watt KD, Pedersen RA, Kremers WK, Heimbach JK, Sanchez W, Gores GJ. Long-term
probability of and mortality from de novo malignancy after liver transplantation.
France, for his review of the manuscript. Medical writing support for Gastroenterology 2009;137:20107.
the authors was funded by Novartis Pharma SAS. [30] Faivre S, Raymond E. Mechanism of action of rapalogues: the antiangiogenic hy-
pothesis. Expert Opin Investig Drugs 2008;17:161921.
[31] Tanaka S, Arii S. Molecular targeted therapies in hepatocellular carcinoma. Semin
References Oncol 2012;39:48692.
[32] Coutte L, Dreyer C, Sablin MP, Faivre S, Raymond E. PI3K-AKT-mTOR pathway and
[1] Fantus D, Thomson AW. Evolving perspectives of mTOR complexes in immunity cancer. Bull Cancer 2012;99:17380.
and transplantation. Am J Transplant 2015;15:891902. [33] Anandappa G, Hollingdale A, Eisen T. Everolimus a new approach in the treat-
[2] Certican (everolimus). Summary of Product Characteristics. European Medicines ment of renal cell carcinoma. Cancer Manag Res 2010;2:6170.
Agency; 2013. [34] Vinayak S, Carlson RW. mTOR inhibitors in the treatment of breast cancer. Oncolo-
[3] Lamoureux F, Picard N, Boussera B, Sauvage FL, Marquet P. Sirolimus and everoli- gy (Williston Park) 2013;27:3844 [46, 48 passim].
mus intestinal absorption and interaction with calcineurin inhibitors: a differential [35] Liu E, Marincola P, Oberg K. Everolimus in the treatment of patients with advanced
effect between cyclosporine and tacrolimus. Fundam Clin Pharmacol 2012;26: pancreatic neuroendocrine tumors: latest ndings and interpretations. Ther Adv
46372. Gastroenterol 2013;6:4129.
[4] Kirchner GI, Meier-Wiedenbach I, Manns MP. Clinical pharmacokinetics of everoli- [36] Bissler JJ, Kingswood JC, Radzikowska E, et al. Everolimus for angiomyolipoma asso-
mus. Clin Pharmacokinet 2004;43:8395. ciated with tuberous sclerosis complex or sporadic lymphangioleiomyomatosis
[5] Gurk-Turner C, Manitpisitkul W, Cooper M. A comprehensive review of everolimus (EXIST-2): a multicentre, randomised, double-blind, placebo-controlled trial. Lan-
clinical reports: a new mammalian target of rapamycin inhibitor. Transplantation cet 2013;381:81724.
2012;94:65968. [37] Lebwohl D, Thomas G, Lane HA, et al. Research and innovation in the development
[6] Webster AC, Lee VW, Chapman JR, Craig JC. Target of rapamycin inhibitors of everolimus for oncology. Expert Opin Drug Discov 2011;6:32338.
(sirolimus and everolimus) for primary immunosuppression of kidney transplant [38] Kauffman HM, Cherikh WS, Cheng Y, Hanto DW, Kahan BD. Maintenance immuno-
recipients: a systematic review and meta-analysis of randomized trials. Transplan- suppression with target-of-rapamycin inhibitors is associated with a reduced inci-
tation 2006;81:123448. dence of de novo malignancies. Transplantation 2005;80:8839.
[7] Manito N, Delgado JF, Crespo-Leiro MG, et al. Clinical recommendations for the use [39] Campistol JM, Eris J, Oberbauer R, et al. Sirolimus therapy after early cyclosporine
of everolimus in heart transplantation. Transplant Rev (Orlando) 2010;24:12942. withdrawal reduces the risk for cancer in adult renal transplantation. J Am Soc
[8] Zuckermann A, Wang SS, Epailly E, et al. Everolimus immunosuppression in de Nephrol 2006;17:5819.
novo heart transplant recipients: what does the evidence tell us now? Transplant [40] Gutirrez-Dalmau A, Campistol JM. The role of proliferation signal inhibitors in
Rev (Orlando) 2013;27:7684. post-transplant malignancies. Nephrol Dial Transplant 2007;22:i116.
[9] De Simone P, Nevens F, De Carlis L, et al. Everolimus with reduced tacrolimus im- [41] De Simone P, Carrai P, Coletti L, Ghinol D, Campani D, Filipponi F. Everolimus-
proves renal function in de novo liver transplant recipients: a randomized con- based immunosuppression is associated with a reduced risk of new-onset malig-
trolled trial. Am J Transplant 2012;12:300820. nancies after liver transplantation. Am J Transplant 2015;15 [Abstract 1284].
[10] Saliba F, De Simone P, Nevens F, et al. Renal function at two years in liver transplant [42] Carenco C, Assenat E, Faure S, et al. Tacrolimus and the risk of solid cancers after
patients receiving everolimus: results of a randomized, multicenter study. Am J liver transplant: a dose effect relationship. Am J Transplant 2015;15:67886.
Transplant 2013;13:173445. [43] Rodrguez-Perlvarez M, De la Mata M, Burroughs AK. Liver transplantation: im-
[11] Fischer L, Klempnauer J, Beckebaum S, et al. A randomized, controlled study to as- munosuppression and oncology. Curr Opin Organ Transplant 2014;19:25360.
sess the conversion from calcineurin-inhibitors to everolimus after liver transplan- [44] Mazzaferro V, Llovet JM, Miceli R, et al, Metroticket Investigator Study Group.
tation PROTECT. Am J Transplant 2012;12:185565. Predicting survival after liver transplantation in patients with hepatocellular

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003
10 J. Dumortier et al. / Transplantation Reviews xxx (2016) xxxxxx

carcinoma beyond the Milan criteria: a retrospective, exploratory analysis. Lancet [74] Hinchey J, Chaves C, Appignani B, et al. A reversible posterior leukoencephalopathy
Oncol 2009;10:3543. syndrome. N Engl J Med 1996;334:494500.
[45] Rodriguez-Perlvarez M, Tsochatzis E, Naveas MC, et al. Reduced exposure to cal- [75] Vizzini G, Asaro M, Miraglia R, et al. Changing picture of central nervous system
cineurin inhibitors early after liver transplantation prevents recurrence of hepato- complications in liver transplant recipients. Liver Transpl 2011;17:127985.
cellular carcinoma. J Hepatol 2013;59:11939. [76] Senzolo M, Ferronato C, Burra P. Neurologic complications after solid organ trans-
[46] Vivarelli M, Cucchetti A, La Barba G, et al. Liver transplantation for hepatocellular plantation. Transpl Int 2009;22:26978.
carcinoma under calcineurin inhibitors: reassessment of risk factors for tumor re- [77] Morard I, Dumortier J, Spahr L, et al. Conversion to sirolimus-based immunosup-
currence. Ann Surg 2008;248:85762. pression in maintenance liver transplantation patients. Liver Transpl 2007;13:
[47] Decaens T, Roudot-Thoraval F, Bresson-Hadni S, et al. Role of immunosuppression 65864.
and tumor differentiation in predicting recurrence after liver transplantation for [78] Finn RS. Current and future treatment strategies for patients with advanced hepa-
hepatocellular carcinoma: a multicenter study of 412 patients. World J tocellular carcinoma: role of mTOR inhibition. Liver Cancer 2012;1:24756.
Gastroenterol 2006;12:731925. [79] De Simone P, Crocetti L, Pezzati D, et al. Efcacy and safety of combination therapy
[48] Duvoux C, Toso C. mTOR inhibitor therapy: Does it prevent HCC recurrence after with everolimus and sorafenib for recurrence of hepatocellular carcinoma after
liver transplantation? Transplant Rev 2015;29:16874. liver transplantation. Transplant Proc 2014;46:2414.
[49] Matter MS, Decaens T, Andersen JB, Thorgeirsson SS. Targeting the mTOR pathway [80] Bilbao I, Sapisochin G, Dopazo C, et al. Indications and management of everolimus
in hepatocellular carcinoma: current state and future trends. J Hepatol 2014;60: after liver transplantation. Transplant Proc 2009;41:21726.
85565. [81] Gomez-Camarero J, Salcedo M, Rincon D, et al. Use of everolimus as a rescue immu-
[50] Schumacher G, Oidtmann M, Rueggeberg A, et al. Sirolimus inhibits growth of nosuppressive therapy in liver transplant patients with neoplasms. Transplantation
human hepatoma cells alone or combined with tacrolimus, while tacrolimus pro- 2007;84:78691.
motes cell growth. World J Gastroenterol 2005;11:14205. [82] Jimnez-Romero C, Manrique A, Marqus E, et al. Switching to sirolimus monother-
[51] Junge G, Saliba F, De Simone P, et al. Everolimus impact on hepatocellular carcino- apy for de novo tumors after liver transplantation. A preliminary experience.
ma recurrence after liver transplantation 12, 24 and 36 months data from 719 Hepato-Gastroenterology 2011;58:11521.
LTX recipients. Am J Transplant 2014;14:694. [83] Garcia VD, Bonamigo Filho JL, Neumann J, et al. Rituximab in association with
[52] Villanueva A, Chiang DY, Newell P, et al. Pivotal role of mTOR signaling in hepato- rapamycin for post-transplant lymphoproliferative disease treatment. Transpl Int
cellular carcinoma. Gastroenterology 2008;135:197283 [83 e111]. 2003;16:2026.
[53] Sahin F, Kannangai R, Adegbola O, Wang J, Su G, Torbenson M. mTOR and P70 S6 [84] Riva G, Luppi M, Barozzi P, Forghieri F, Potenza L. How I treat HHV8/KSHV-related
kinase expression in primary liver neoplasms. Clin Cancer Res 2004;10:84215. diseases in posttransplant patients. Blood 2012;120:41509.
[54] Sato K, Mori M. Evolving molecular mechanism-based strategies for control of he- [85] Ducroux E, Boillot O, Ocampo MA, et al. Skin cancers after liver transplantation: ret-
patocellular carcinoma. Curr Med Chem 2011;18:437588. rospective single-center study on 371 recipients. Transplantation 2014;98:33540.
[55] Huynh H, Chow KH, Soo KC, et al. RAD001 (everolimus) inhibits tumour growth in [86] Euvrard S, Morelon E, Rostaing L, et al. Sirolimus and secondary skin-cancer pre-
xenograft models of human hepatocellular carcinoma. J Cell Mol Med 2009;13: vention in kidney transplantation. N Engl J Med 2012;367:32939.
137180. [87] Kaplan B, Qazi Y, Wellen JR. Strategies for the management of adverse events asso-
[56] Toso C, Merani S, Bigam DL, Shapiro AM, Kneteman NM. Sirolimus-based immuno- ciated with mTOR inhibitors. Transplant Rev (Orlando) 2014;28:12633.
suppression is associated with increased survival after liver transplantation for he- [88] Sepulveda A, Scatton O, Tranchart H, et al. Split liver transplantation using extend-
patocellular carcinoma. Hepatology 2010;51:123743. ed right grafts: the natural history of segment 4 and its impact on early postoper-
[57] Cholongitas E, Mamou C, Rodrguez-Castro KI, Burra P. Mammalian target of ative outcomes. Liver Transpl 2012;18:41322.
rapamycin inhibitors are associated with lower rates of hepatocellular carcinoma [89] Gharbi C, Gueutin V, Izzedine H. Oedema, solid organ transplantation and mamma-
recurrence after liver transplantation: a systematic review. Transpl Int 2014;27: lian target of rapamycin inhibitor/proliferation signal inhibitors (mTOR-I/PSIs). Clin
103949. Kidney J 2014;7:11520.
[58] Ferreiro AO, Vazquez-Milln MA, Lpez FS, Gutirrez MG, Diaz SP, Patio MJ. [90] Buron F, Malvezzi P, Villar E, et al. Proling sirolimus-induced inammatory syn-
Everolimus-based immunosuppression in patients with hepatocellular carcinoma drome: a prospective tricentric observational study. PLoS One 2013;8:e53078.
at high risk of recurrence after liver transplantation: a case series. Transplant [91] Holdaas H, Potena L, Saliba F. mTOR inhibitors and dyslipidemia in transplant recip-
Proc 2014;46:3496501. ients: a cause for concern? Transplant Rev (Orlando) 2015;29:93102.
[59] Toso C, Mentha G, Majno P. Liver transplantation for hepatocellular carcinoma: ve [92] Calmus Y, Durrbach A. Everolimus de novo in liver transplantation. Gastroenterol
steps to prevent recurrence. Am J Transplant 2011;11:20315. Clin Biol 2009;33:S24752.
[60] Saliba F, Nevens F. Progression of liver brosis in HCV-positive liver transplant re- [93] Diekmann F, Andrs A, Oppenheimer F. mTOR inhibitor-associated proteinuria in
cipients randomized to everolimus with reduced calcineurin inhibitor (CNI) thera- kidney transplant recipients. Transplant Rev (Orlando) 2012;26:279.
py or a standard CNI regimen. Transpl Int 2015;28:3734. [94] Lopez P, Kohler S, Dimri S. Interstitial lung disease associated with mTOR inhibitors
[61] Wagner D, Kniepeiss D, Schaffellner S, et al. Sirolimus has a potential to inuent in solid organ transplant recipients: results from a large phase III clinical trial pro-
viral recurrence in HCV positive liver transplant candidates. Int Immunopharmacol gram of everolimus and review of the literature. J Transplant 2014;2014:305931.
2010;10:9903. [95] van der Vliet JA, Willems MC, de Man BM, Lomme RM, Hendriks T. Everolimus in-
[62] Asthana S, Toso C, Meeberg G, et al. The impact of sirolimus on hepatitis C recur- terferes with healing of experimental intestinal anastomoses. Transplantation
rence after liver transplantation. Can J Gastroenterol 2011;25:2834. 2006;82:147783.
[63] McKenna GJ, Trotter JF, Klintmalm E, et al. Limiting hepatitis C virus progression in [96] Cooper M, Wiseman AC, Zibari G, et al. Wound events in kidney transplant patients
liver transplant recipients using sirolimus-based immunosuppression. Am J Trans- receiving de novo everolimus: a pooled analysis of three randomized controlled tri-
plant 2011;11:237987. als. Clin Transpl 2013;27:E62535.
[64] Liverpool Drugs Interactions website. Available at: www.hep-druginteractions.org [97] Schwarz L, Cauchy F, Conti F, et al. Preliminary report of major surgery in liver
[Accessed 20 May 2015] transplant recipients receiving m-TOR inhibitors without therapeutic discontinua-
[65] Saliba F, Dharancy S, Lorho R, et al. Conversion to everolimus in maintenance liver tion. World J Surg 2014;38:31938.
transplant patients: a multicenter, retrospective analysis. Liver Transpl 2011;17: [98] Pallet N, Legendre C. Adverse events associated with mTOR inhibitors. Expert Opin
90513. Drug Saf 2013;12:17786.
[66] Saliba F, Dharancy S, Lorho R, et al. Everolimus (EVL) after liver transplantation [99] Huyghe E, Zairi A, Nohra J, Kamar N, Plante P, Rostaing L. Gonadal impact of target
(LT): a retrospective multicenter study evaluating modalities, efcacy and safety of rapamycin inhibitors (sirolimus and everolimus) in male patients: an overview.
of conversion to EVL (EVEROLIVER study). Transplantation 2010;90:643 [Suppl.]. Transpl Int 2007;20:30511.
[67] Bilbao I, Dopazo C, Lazaro J, et al. Multiple indications for everolimus after liver [100] Carta P, Zanazzi M, Minetti EE. Unplanned pregnancies in kidney transplanted patients
transplantation in current clinical practice. World J Transplant 2014;4:12232. treated with everolimus: three case reports. Transpl Int 2015;28:3702.
[68] Vallin M, Guillaud O, Morard I, et al. Tolerability of everolimus-based immunosup- [101] Rostaing L, Christiaans MH, Kovarik JM, Pascual J. The pharmacokinetics of everoli-
pression in maintenance liver transplant recipients. Clin Transpl 2011;25:6609. mus in de novo kidney transplant patients receiving tacrolimus: an analysis from
[69] Thimonier E, Guillaud O, Walter T, et al. Conversion to everolimus dramatically im- the randomized ASSET study. Ann Transplant 2014;19:33745.
proves the prognosis of de novo malignancies after liver transplantation for alcohol- [102] Kovarik JM, Curtis JJ, Hricik DE, Pescovitz MD, Scantlebury V, Vasquez A. Differential
ic liver disease. Clin Transpl 2014;28:133948. pharmacokinetic interaction of tacrolimus and cyclosporine on everolimus. Trans-
[70] Alegre C, Jimnez C, Manrique A, et al. Everolimus monotherapy or combined therapy plant Proc 2006;38:34568.
in liver transplantation: indications and results. Transplant Proc 2013;45:19714. [103] Nashan B, Schemmer P, Braun F, Dworak M, Wimmer P, Schlitt H. Evaluating the
[71] Prez T, Segovia R, Castro L, Roblero JP, Estela R. Conversion to everolimus in liver efcacy, safety and evolution of renal function with early initiation of
transplant patients with renal dysfunction. Transplant Proc 2011;43:230710. everolimus-facilitated tacrolimus reduction in de novo liver transplant recipients:
[72] Castroagudin JF, Molina E, Romero R, Otero E, Tom S, Varo E. Improvement of renal study protocol for a randomized controlled trial. Trials 2015;16:118.
function after the switch from a calcineurin inhibitor to everolimus in liver trans- [104] Rostaing L, Hertig A, Albano L, et al, on behalf of the CERTITEM Study Group. Fibro-
plant recipients with chronic renal dysfunction. Liver Transpl 2009;15:17927. sis progression according to epithelial-mesenchymal transition prole: a random-
[73] Flechner SM, Kobashigawa J, Klintmalm G. Calcineurin inhibitor-sparing regimens ized trial of everolimus versus CsA. Am J Transplant 2015;15:130212.
in solid organ transplantation: focus on improving renal function and nephrotoxi- [105] Llovet JM. Liver cancer: time to evolve trial design after everolimus failure. Nat Rev
city. Clin Transpl 2008;22:115. Clin Oncol 2014;11:5067.

Please cite this article as: Dumortier J, et al, Use of everolimus in liver transplantation: The French experience, Transplant Rev (2016), http://
dx.doi.org/10.1016/j.trre.2015.12.003

You might also like