18
Everolimus (Certican) in renal transplantation: a review of clinical trial data, current usage, and future directions Julio Pascual a, 4 , Ioannis N. Boletis b , Josep M. Campistol c a Servicio de Nefrologia, Hospital Ramo ´n y Cajal, 28034 Madrid, Spain b Department of Nephrology and Transplantation, Laiko Hospital, 15562 Athens, Greece c Servei de Nefrologia i Trasplantament Renal, Hospital Clı ´nic de Barcelona, Universitat de Barcelona, 08036 Barcelona, Spain Abstract The efficacy and tolerability of everolimus have been demonstrated in a number of clinical trials, and there is also an increasing body of clinical experience. The efficacy of everolimus after renal transplantation is at least equivalent to that of mycophenolate mofetil. Studies combining everolimus with full- or reduced-dose cyclosporine (CsA) have shown that CsA exposure can be minimized, without increasing the risk of acute rejection, particularly when combined with therapeutic drug monitoring. A role for everolimus in regimens involving elimination of calcineurin inhibitors is currently being investigated. Everolimus with significantly reduced-dose CsA has not been shown to enhance CsA-related nephrotoxicity. Adverse events seen in trials of everolimus are generally class-specific and include edema, arthralgia, dyslipidemia, impaired wound healing, and proteinuria. A low incidence of malignancy has been observed with everolimus, and studies are ongoing to examine its antitumor effects in the treatment of certain malignancies. It seems likely that everolimus will continue to play a role in the development of reduced-exposure calcineurin inhibitor regimens and has considerable potential to improve outcomes for transplant recipients, focused perhaps on bold-for-oldQ transplant recipients and patients at high risk of poor graft function or malignancy. This review considers the available data on the clinical application of everolimus and identifies current and future strategies for improving outcomes after renal transplantation. D 2006 Elsevier Inc. All rights reserved. 1. Introduction Survival of renal organ transplant recipients has improved greatly over the years, particularly since the introduction of calcineurin inhibitors (CNIs), such as cyclosporine (CsA), and purine biosynthesis inhibitors (eg, mycophenolic acids [MPAs]). Renal graft survival rates are now in the order of 90% at 1-year posttransplant [1], but ensuring very long-term graft survival (ie, up to 10 years) remains a challenge. Chronic allograft nephrop- athy (CAN), characterized by interstitial fibrosis and tubular atrophy [2], is the main cause of renal graft loss, although patient death is also an important cause of loss of otherwise functioning grafts. Risk factors for the develop- ment of CAN include acute rejection, use of CNIs, cytomegalovirus (CMV) infection, and comorbidities such as hypertension and hyperlipidemia [3]. A recent longitu- dinal study charted the course of CAN in 120 renal transplant recipients [4]. Chronic allograft nephropathy was shown to be a progressive, time-dependent process, with much tubulointerstitial damage occurring within 1 to 2 years of transplantation. Later damage was characterized by progressive arteriolar hyalinosis, ischemic glomerulo- sclerosis, and further interstitial fibrosis associated with CNI toxicity [4], highlighting the difficult balance that physicians must achieve with CNIs, the mainstay of immunosuppressive therapy. Despite their efficacy, these agents were almost universally associated with nephrotoxic effects at 10-year follow-up. Further improvements in renal graft survival will depend on the development of immu- nosuppressive regimens that can prevent CAN and do so without inducing nephrotoxic effects. A new class of immunosuppressant agents, proliferation signal inhibitors (PSIs; also known as mammalian target of rapamycin inhibitors), sirolimus (Rapamune, Wyeth Pharmaceuticals, Madison, NJ), and everolimus (Certican, Novartis Pharma AG, Basel, Switzerland) has potential in this respect. This review will concentrate on the clinical trial data for everolimus and its use in clinical practice. 0955-470X/$ – see front matter D 2006 Elsevier Inc. All rights reserved. doi:10.1016/j.trre.2005.10.005 4 Corresponding author. Tel.: +34 913368018. E-mail address: [email protected] (J. Pascual). Transplantation Reviews 20 (2006) 1 – 18 www.elsevier.com/locate/trre

Everolimus (Certican) in renal transplantation: a review of clinical trial data, current usage, and future directions

Embed Size (px)

Citation preview

  • sp

    an

    BoRamo

    ntatio

    de B

    ted in

    ntatio

    ) ha

    peut

    Ever

    f eve

    dinal study charted the course of CAN in 120 renalinhibitors), sirolimus (Rapamune, Wyeth Pharmaceuticals,

    Madison, NJ), and everolimus (Certican, Novartis Pharma

    AG, Basel, Switzerland) has potential in this respect. This

    Transplantation Reviewsdyslipidemia, impaired wound healing, and proteinuria. A low incidence of malignancy has been observed with everolimus, and studies are

    ongoing to examine its antitumor effects in the treatment of certain malignancies. It seems likely that everolimus will continue to play a role

    in the development of reduced-exposure calcineurin inhibitor regimens and has considerable potential to improve outcomes for transplant

    recipients, focused perhaps on bold-for-oldQ transplant recipients and patients at high risk of poor graft function or malignancy. This reviewconsiders the available data on the clinical application of everolimus and identifies current and future strategies for improving outcomes after

    renal transplantation.

    D 2006 Elsevier Inc. All rights reserved.

    1. Introduction

    Survival of renal organ transplant recipients has

    improved greatly over the years, particularly since the

    introduction of calcineurin inhibitors (CNIs), such as

    cyclosporine (CsA), and purine biosynthesis inhibitors

    (eg, mycophenolic acids [MPAs]). Renal graft survival

    rates are now in the order of 90% at 1-year posttransplant

    [1], but ensuring very long-term graft survival (ie, up to

    10 years) remains a challenge. Chronic allograft nephrop-

    athy (CAN), characterized by interstitial fibrosis and

    tubular atrophy [2], is the main cause of renal graft loss,

    although patient death is also an important cause of loss of

    otherwise functioning grafts. Risk factors for the develop-

    ment of CAN include acute rejection, use of CNIs,

    cytomegalovirus (CMV) infection, and comorbidities such

    as hypertension and hyperlipidemia [3]. A recent longitu-

    transplant recipients [4]. Chronic allograft nephropathy

    was shown to be a progressive, time-dependent process,

    with much tubulointerstitial damage occurring within 1 to

    2 years of transplantation. Later damage was characterized

    by progressive arteriolar hyalinosis, ischemic glomerulo-

    sclerosis, and further interstitial fibrosis associated with

    CNI toxicity [4], highlighting the difficult balance that

    physicians must achieve with CNIs, the mainstay of

    immunosuppressive therapy. Despite their efficacy, these

    agents were almost universally associated with nephrotoxic

    effects at 10-year follow-up. Further improvements in renal

    graft survival will depend on the development of immu-

    nosuppressive regimens that can prevent CAN and do so

    without inducing nephrotoxic effects. A new class of

    immunosuppressant agents, proliferation signal inhibitors

    (PSIs; also known as mammalian target of rapamycinEverolimus (Certican) in renal tran

    data, current usage,

    Julio Pascuala,4, Ioannis N.aServicio de Nefrologia, Hospital

    bDepartment of Nephrology and TransplacServei de Nefrologia i Trasplantament Renal, Hospital Clnic

    Abstract

    The efficacy and tolerability of everolimus have been demonstra

    clinical experience. The efficacy of everolimus after renal transpla

    combining everolimus with full- or reduced-dose cyclosporine (CsA

    the risk of acute rejection, particularly when combined with thera

    elimination of calcineurin inhibitors is currently being investigated.

    enhance CsA-related nephrotoxicity. Adverse events seen in trials o0955-470X/$ see front matter D 2006 Elsevier Inc. All rights reserved.

    doi:10.1016/j.trre.2005.10.005

    E-mail address: [email protected] (J. Pascual).lantation: a review of clinical trial

    d future directions

    letisb, Josep M. Campistolc

    n y Cajal, 28034 Madrid, Spain

    n, Laiko Hospital, 15562 Athens, Greece

    arcelona, Universitat de Barcelona, 08036 Barcelona, Spain

    a number of clinical trials, and there is also an increasing body of

    n is at least equivalent to that of mycophenolate mofetil. Studies

    ve shown that CsA exposure can be minimized, without increasing

    ic drug monitoring. A role for everolimus in regimens involving

    olimus with significantly reduced-dose CsA has not been shown to

    rolimus are generally class-specific and include edema, arthralgia,

    20 (2006) 118

    www.elsevier.com/locate/trrereview will concentrate on the clinical trial data for4 Corresponding author. Tel.: +34 913368018.

    everolimus and its use in clinical practice.

  • 2. Everolimusclinical development and efficacy in de

    novo renal transplantation

    2.1. Mechanism of action

    Everolimus has a mode of action that is distinct from

    CNIs, which inhibit the transcription of early T-cellspecific

    genes, thus reducing the production of T-cell growth factors

    such as interleukin (IL) 2 [5]. Everolimus acts at a later

    stage of the cell cycle, blocking the proliferation signal

    provided by these growth factors and preventing cells from

    entering the S phase [5]. The antiproliferative actions of

    everolimus are not limited to the immune system; it also

    inhibits growth factordriven cell proliferation in general

    (eg, reducing vascular smooth muscle cell proliferation) [5].

    The antiproliferative effects of everolimus also prevent

    vascular remodeling [6], a key component of progressive

    (44% and 50%, respectively), with the most frequent

    adverse events being headache in the everolimus groups

    and dizziness among those receiving placebo. Mean

    changes in laboratory parameters did not differ significantly

    between everolimus and placebo groups. Importantly,

    administration of everolimus in single doses did not appear

    to affect the steady-state pharmacokinetics of CsA in these

    individuals [14]. A further study in healthy individuals

    evaluated coadministration of everolimus with 2 different

    CsA formulationsa microemulsion (Neoral, Novartis

    Pharma AG) and an oral solution (Sandimmune, Novartis

    Pharma AG) [15]. Both CsA formulations were observed to

    increase systemic exposure (area under the concentration-

    time curve [AUC]) to everolimus, but the percentage or

    increase was significantly greater for the microemulsion

    formulation (P = .008). A 2- to 3-fold decrease in

    raft dy

    ermis

    J. Pascual et al. / Transplantation Reviews 20 (2006) 1182allograft dysfunction (Fig. 1) [7].

    Preclinical studies have demonstrated the immunosup-

    pressive actions of everolimus in animal models of renal

    transplantation [8-11]. Moreover, such models provide

    evidence for synergistic activity between everolimus and

    CsA, with the 2 combined agents offering more potent

    immunosuppression than either one alone [8,12]. Animal

    models have also been used to demonstrate the anti-

    proliferative effect of everolimus on nonimmune cells.

    For example, everolimus was associated with delayed

    progression of CAN in a rat model of renal transplan-

    tation because of antiproliferative or apoptosis-enhancing

    effects [13].

    2.2. Early clinical development

    In the first study of everolimus in humans, 54 stable renal

    transplant recipients split into 6 groups received either a

    single dose of everolimus, ranging from 0.25 to 25 mg or

    placebo in addition to CsA [14]. The safety of each dose

    was monitored for 11 days before the next group of

    individuals received a higher dose. All doses of everolimus

    were well tolerated. Similar proportions of patients receiv-

    ing everolimus and placebo had at least 1 adverse event

    Fig. 1. Vascular remodeling in transplanted organs, a key component of allog

    to cytokine release and up-regulation of growth factors. Reproduced with peverolimus exposure can therefore be expected after

    withdrawal of CsA from immunosuppressant regimens.

    This pharmacokinetic interaction may be due to direct

    competition between everolimus and CsA because both

    agents undergo metabolism by CYP3A4 and are substrates

    of P-glycoprotein. Conversely, if CsA is added to an

    everolimus-containing regimen, an average 3-fold increase

    in systemic exposure to everolimus is observed [16].

    After administration of single doses of everolimus

    (0.2515 mg) to 7 renal transplant recipients also receiving

    standard CsA immunosuppression, the pharmacokinetics of

    CsA were again unaffected by everolimus administration

    [17]. The mean maximal plasma concentration (Cmax) of

    everolimus (dose normalized to 1 mg everolimus) was 7.9F2.7 lg/L, whereas the mean time to maximum plasmaconcentration (Tmax) was 1.5F 0.9 hours. In a larger phase Istudy, 24 renal transplant recipients were given everolimus

    0.75, 2.5, or 7.5 mg/d, or placebo for 4 weeks [18]. Mean

    initial Tmax varied from 1.3 F 0.3 to 1.8 F 0.5 hours forthe 3 everolimus groups, and steady state was reached

    within 4 days. The mean elimination half-life was 19.2 F3.4, 18.1 F 7.6, and 16.0 F 5.6 hours for the 0.75, 2.5,and 7.5-mg groups, respectively. The pharmacokinetics of

    sfunction. Continued immune and endothelial activation posttransplant lead

    sion from Neumayer [7].

  • everolimus showed dose-exposure proportionality and a

    good correlation between trough and AUC concentrations.

    The most frequently occurring adverse event was an

    increased incidence of infection. Thrombocytopenia and

    hyperlipidemia occurred in a dose-dependent manner.

    In a phase II trial, 101 de novo renal transplant

    recipients receiving everolimus 1.0, 2.0, or 4.0 mg/d were

    monitored for 1 year posttransplant [19]. Steady-state

    pharmacokinetics was achieved on or before day 7. Cmaxand systemic exposure (AUC) were proportional to ever-

    olimus dose, with AUC being stable throughout follow-up.

    Pharmacokinetic parameters associated with CsA treatment

    did not differ with administration of everolimus at any

    dose. The incidence of acute rejection was analyzed in 103

    de novo renal transplant recipients who were randomized

    to receive everolimus 1.0, 2.0, or 4.0 mg/d in addition to

    CsA and corticosteroids [20]. Over the first 6 months of

    treatment, 32.4%, 14.7%, and 25.7% of patients in the

    respective treatment groups experienced biopsy-proven

    acute rejection (BPAR). Compared with the everolimus

    1.0 mg/d group, the incidence of moderate and severe

    BPAR was significantly lower in the 2.0 and 4.0 mg/d

    groups (P = .002 and .006, respectively). Everolimus was

    generally well tolerated, although viral and fungal infec-

    tions were more common in patients receiving the highest

    dose. As in previous studies, blood lipid levels were

    elevated during everolimus treatment, requiring lipid-

    2.3. Clinical efficacy studies

    Clinical development of everolimus in de novo renal

    transplantation was supported by a long-term phase II study

    and 4 major phase III clinical trials (Table 1). A 3-year

    phase II trial (study B156) compared the efficacy and

    tolerability of everolimus in combination with full- and

    reduced-dose CsA [21]. Two 3-year phase III trials (studies

    B201 and B251) were conducted to establish whether

    everolimus was similar in efficacy to mycophenolate mofetil

    (MMF) in renal transplantation when both agents were

    combined with full-dose CsA and corticosteroids [22-24].

    Subsequently, the efficacy of everolimus in combination

    with reduced-dose CsA was investigated with the aim of

    establishing immunosuppressive regimens with lower CNI-

    induced nephrotoxicity (studies A2306 and A2307) [25].

    2.3.1. Everolimus vs MMF with full-dose CsA

    The large-scale trials, studies B201 [22,23] and B251

    [24], were of a similar design, with de novo renal transplant

    recipients being randomized to receive fixed doses of

    everolimus 1.5 mg/d, everolimus 3.0 mg/d, or MMF

    2.0 g/d in a blinded fashion for 1 year, followed by 2 years

    of open-label treatment. In addition, all patients received

    full-dose CsA microemulsion (Neoral) and corticosteroids.

    The primary objective in both cases was to compare the

    incidence of an efficacy composite end pointefficacy

    le)

    le)

    re CsA

    J. Pascual et al. / Transplantation Reviews 20 (2006) 118 3lowering agents.

    Table 1

    Clinical trials for the development of everolimus in renal transplantation

    Study Study design Study duration

    B156 Phase IIrandomized,

    open-label, parallel-group

    3 y

    B201 Fixed-dose everolimus 3 y

    Phase IIIrandomized,

    double-blind, double-dummy,

    parallel-group

    Open study years 23

    B251 Fixed-dose everolimus 3 y

    Phase IIIrandomized,

    double-blind,

    double-dummy,

    parallel-group

    Open study years 23

    A2306 Concentration-controlled

    everolimus

    1 y (6-mo data availab

    Phase IIIrandomized,

    open-label, parallel-group

    A2307 Concentration-controlled

    everolimus

    1 y (6-mo data availab

    Phase IIIrandomized,

    open-label, parallel-group

    a In combination with basiliximab and corticosteroids.b In combination with standard-dose CsA and corticosteroids.c In combination with standard-dose CsA and prednisone.d In combination with corticosteroids and reduced-exposure CsA.e In combination with basiliximab, corticosteroids, and reduced-exposufailurein each treatment group at 12 or 36 months

    Treatment Patients (n) References

    Everolimus 3.0 mg/d +

    full-dose CsAa53 Nashan et al [21]

    Everolimus 3.0 mg/d +

    reduced-dose CsAa58

    Everolimus 1.5 mg/db 194 Vtko et al [22,23]

    Everolimus 3.0 mg/db

    MMF 2.0 g/db198

    196

    Everolimus 1.5 mg/dc 193 Lorber et al [24]

    Everolimus 3.0 mg/dc

    MMF 2.0 g/dc194

    196

    Everolimus 1.5 mg/dd

    Everolimus 3.0 mg/dd112

    125

    Vtko et al [25]

    Everolimus 1.5 mg/de

    Everolimus 3.0 mg/de117

    139

    Vtko et al [25]

    .

  • posttransplant. Efficacy failure was defined as the incidence

    of BPAR, graft loss, death, or loss to follow-up. Secondary

    end points included allograft and patient survival, as well as

    the incidence of acute rejection and safety parameters.

    The incidences of efficacy failure, BPAR, graft loss, and

    death at 12 and 36 months of follow-up in studies B201 and

    B251 are shown in Fig. 2 and were similar among patients

    randomized to MMF and everolimus 1.5 or 3.0 mg/d.

    Furthermore, the therapeutic equivalence of everolimus and

    MMF was maintained in the long term, as there were no

    significant differences between efficacy end points in either

    trial at 36 months of follow-up [23,24]. A significant

    difference between the treatment arms was noted in study

    B251 [24], in which the incidence of antibody-treated acute

    rejection was significantly lower among patients receiving

    everolimus 1.5 mg/d than among those receiving MMF at

    12 months (7.8% vs 16.3%; P = .01) and 36 months of

    follow-up (9.8% vs 18.4%, respectively; P = .014). In both

    studies, most graft loss occurred within the first 12 months

    of treatment. In study B251, reasons for graft loss included

    infection, acute and chronic rejection, kidney infarction, and

    recurrence of original disease [24].

    The interactions noted in studies B201 and B251

    highlighted that therapeutic drug monitoring (TDM) could

    be beneficial for patients receiving everolimus and CsA.

    Analysis of data from 779 patients in the 2 trials showed that

    everolimus trough blood levels of 3 ng/mL or more were

    necessary to gain most clinical benefit from the agent [26].

    Risk of BPAR was 3.4-fold higher for patients with

    everolimus trough blood levels less than 3 ng/mL compared

    B201

    J. Pascual et al. / Transplantation Reviews 20 (2006) 1184Fig. 2. Efficacy analyses at 12 and 36 months of follow-up in clinical trials

    graft loss, death, loss to follow-up [22-24].and B251 (intent-to-treat population). Primary efficacy is defined as BPAR,

  • J. Pascual et al. / Transplantation Reviews 20 (2006) 118 5with patients whose trough blood levels were in the range 3

    to 8 ng/mL (P b .0001) (Fig. 3) [26]. There was a trendtoward even lower rates of BPAR in patients with ever-

    olimus trough blood levels greater than 8 ng/mL, but this

    finding should be interpreted cautiously given the small

    number of patients in this group. It was estimated that after

    initiation of everolimus at a dose of 1.5 mg/d, 2 TDM-led

    dose adjustments would be sufficient for 84% of patients to

    achieve everolimus trough blood levels in excess of

    3 ng/mL. With respect to renal function, the TDM analysis

    showed that risk of serum creatinine levels of 200 lmol/L ormore increased greatly with increasing CsA trough levels,

    but was affected only slightly by everolimus trough blood

    levels [26].

    Monitoring of serum creatinine levels in studies B201

    and B251 showed that patients receiving everolimus

    Fig. 3. Kaplan-Meier plots over the first 6 months of treatment showing the

    percentage of patients free from BPAR at different everolimus trough blood

    levels (n = 779). Reproduced with permission from Lorber et al [26].exhibited higher mean serum creatinine levels than those

    receiving MMF [22-24]. At 12 months of follow-up, a

    protocol amendment was introduced in both trials allowing

    for lower CsA trough blood levels (5075 ng/mL) to be

    targeted in the everolimus groups, provided everolimus

    trough blood levels were at least 3 ng/mL. Mean serum

    creatinine levels subsequently decreased slightly or

    remained stable in everolimus-treated patients, and CsA

    reduction was not accompanied by an increase in BPAR.

    Everolimus itself does not induce renal dysfunction, but

    these findings appeared to indicate that it can potentiate

    CsA-related nephrotoxicity. A similar effect has been noted

    in patients receiving sirolimus [27].

    In addition to efficacy and safety analyses, an economic

    evaluation of treatment was also carried out during study

    B201 [28]. At 12 months of follow-up, mean overall

    treatment costs were US$33 715, US$38 519, and US$36

    509 for the everolimus 1.5 mg, everolimus 3.0 mg, and

    MMF groups, respectively. Differences in cost between the

    treatment groups were not significant.2.3.2. Everolimus combined with full- or reduced-dose CsA

    After protocol amendment, data from studies B201 and

    B251 indicated that everolimus plus reduced-exposure CsA

    could result in improved renal function without increased

    risk of rejection. This is supported by the findings of a

    3-year phase II trial (study 156) in which patients received

    everolimus 3.0 mg/d in combination with basiliximab,

    corticosteroids, and either full- (n = 53) or reduced-dose

    (n = 58) CsA microemulsion [21]. From 3 to 36 months of

    follow-up, target CsA trough blood levels were 125 to

    250 ng/mL in the full-dose CsA group, compared with 50

    to 100 ng/mL in the reduced-dose CsA group. The

    incidence of efficacy failure (BPAR, graft loss, death, or

    loss to follow-up) was significantly lower in the reduced-

    dose CsA group compared with the full-dose CsA group at

    6 (P = .046), 12 (P = .012), and 36 (P = .032) months

    of follow-up [21]. More specifically, BPAR was less

    frequent in the reduced-dose CsA group than in the full-

    dose CsA group at each follow-up time point: 6 months,

    3.4% vs 15.1%; 12 months, 6.9% vs 17.0%; and

    36 months, 12.1% vs 18.9%, respectively.

    Importantly, at 6 and 12 months of follow-up, mean

    serum creatinine levels were numerically lower in patients

    randomized to reduced CsA dosing than in those receiving

    full-dose CsA [21]. The advantage of reduced-dose CsAwas

    more evident in relation to mean creatinine clearance values,

    which were significantly higher in the reduced-dose group

    at 6 (P = .009) and 12 (P = .007) months of follow-up.

    Patients remaining on treatment at 12 months were treated

    according to an amended protocol to optimize their renal

    function: CsA dosing was adjusted to achieve trough blood

    levels of 50 to 75 ng/mL, whereas everolimus dose was

    adjusted to ensure trough blood levels of at least 3 ng/mL

    (in accordance with TDM study findings). After transition to

    the amended protocol, mean serum creatinine levels in the

    full-dose CsA group fell. In patients randomized to reduced-

    dose CsA, mean serum creatinine and creatinine clearance

    values remained stable, reflecting the smaller reduction in

    CsA dose for this group [21].

    Overall, study B156 demonstrated how synergy between

    everolimus and CsA permitted reduced-dose CsA, resulting

    in lower rates of efficacy failure and improved renal

    function compared with everolimus and full-dose CsA [21].

    2.3.3. Concentration-controlled everolimus plus

    reduced-exposure CsA

    Therapeutic use of everolimus in combination with CsA

    was further refined in 2 similarly designed phase III clinical

    trials in which TDM was used to optimize everolimus

    dosing, and the CsA blood levels 2 hours after administra-

    tion (C2 monitoring) was used to guide reduction in CsA

    exposure [25]. In study A2306, concentration-controlled

    everolimus 1.5 or 3.0 mg/d was combined with reduced-

    exposure CsA (given as CsA microemulsion) and cortico-

    steroids; the same approach was used in study A2307

    with the exception that patients also received doses of

  • and A2307) examining the use concentration-controlled everolimus with reduced-

    out basiliximab) Study A2307 (with basiliximab)

    /d Everolimus 3.0 mg/d Everolimus 1.5 mg/d Everolimus 3.0 mg/d

    2) 62 (62) (n = 111) 66 (66) (n = 107) 67 (65) (n = 123)

    2) 62 (65) (n = 125) 64 (68) (n = 117) 65 (65) (n = 139)

    2) 61 (63) (n = 125) 63 (60) (n = 117) 61 (60) (n = 139)

    5) 132 (140) (n = 112) 130 (137) (n = 113) 130 (136) (n = 127)

    2) 126 (134) (n = 125) 128 (132) (n = 117) 126 (132) (n = 139)

    2) 128 (141) (n = 125) 142 (136) (n = 117) 142 (136) (n = 139)

    24/125 (19.2%) 18/117 (15.4%) 27/139 (19.4%)

    32/125 (26%) 23/117 (20%) 34/139 (25%)

    34/125 (27%) 23/117 (20%) 34/139 (25%)

    19/125 (15.2%) 16/117 (13.7%) 21/139 (15.1%)

    24/125 (19%) 16/117 (13.7%) 22/139 (15.8%)

    26/125 (21%) 19/117 (16%) 25/139 (18%)

    J. Pascual et al. / Transplantation Reviews 20 (2006) 1186basiliximab on the day of transplantation and 4 days later.

    Everolimus dose was adjusted to ensure trough blood levels

    of at least 3 ng/mL. In study A2306, CsA C2 level target

    ranges were 1000 to 1400 ng/mL during weeks 0 to 4, 700

    to 900 ng/mL during weeks 5 to 8, 550 to 650 ng/mL during

    weeks 9 to 12, and 350 to 450 ng/mL thereafter. Because of

    the use of induction therapy, target CsA C2 ranges were set

    lower in study A2307: 500 to 700 ng/mL during weeks 0 to

    8 and 350 to 450 ng/mL from week 9 onward.

    The primary end point of the trials was renal function as

    assessed by glomerular filtration rate (GFR) and creatinine

    clearance or serum creatinine [25]. Table 2 shows values for

    GFR and serum creatinine at 6, 12, and 24 months of

    follow-up in both trials [25,29-33]. Serum creatinine values

    were substantially lower than those observed among

    patients receiving everolimus with full-dose CsA in study

    Table 2

    Renal function and efficacy parameters in 2 similarly designed trials (A2306

    exposure CsA in renal transplantation [25,29-33]

    Follow-up period (mo) Study A2306 (with

    Everolimus 1.5 mg

    Renal function

    Median (mean)

    calculated GFR

    (creatinine clearance;

    mL/min)

    6 65 (63) (n = 10

    12 69 (69) (n = 11

    24 68 (66) (n = 11

    Median (mean) serum

    creatinine (lmol/L)6 133 (147) (n = 10

    12 122 (126) (n = 11

    24 129 (133) (n = 11

    Efficacy

    Efficacy failure, n (%) 6 31/112 (27.7%)

    12 31/112 (28%)

    24 34/112 (30%)

    BPAR, n (%) 6 28/112 (25%)

    12 29/112 (26%)

    24 30/112 (27%)B251 [24].

    Efficacy end points are also listed in Table 2. The

    incidence of efficacy failure in both studies was equivalent

    to, or lower than, that in earlier studies in which full doses

    of CsA were used. Efficacy failure occurred more

    frequently in study A2306 compared with study A2307,

    mainly because of higher rates of BPAR in the first

    6 months posttransplant [25]. These data confirm the added

    benefit of IL-2 receptor antagonist induction therapy to

    reduce risk of early BPAR in combination with a lower

    dose of everolimus.

    Infection was the most common adverse event, occurring

    in 61.6% and 56.8% of patients randomized to everolimus

    1.5 or 3.0 mg/d in study A2306 within the first 6 months of

    treatment [25]. Interestingly, CMV disease was not com-

    mon; in study A2306, it occurred in just 0.9% of patients

    receiving everolimus 1.5 mg/d and in 3.2% of patients

    receiving everolimus 3.0 mg/d. In the same trial, thrombo-

    cytopenia occurred in 3.6% of the everolimus 1.5 mg/d

    group and in 8.0% of the everolimus 3.0 mg/d group. Otheradverse events included anemia, which occurred in 18.8%

    and 23.9% of the everolimus 1.5 mg/d groups of studies

    A2306 and A2307, respectively, and lymphocele, the

    incidence of which varied from 6.4% to 15.2% in the

    4 treatment groups. In study A2306, mean total serum

    cholesterol rose from 4.2 and 3.9 mmol/L at baseline to

    6.6 and 6.5 mmol/L at 6 months in the everolimus 1.5 and

    3.0 mg/d groups, respectively [25]. Similar elevations were

    noted in study A2307 with hyperlipidemia generally

    manifesting early in treatment and lipid levels stabilizing

    after 2 to 3 months.

    No upper limit for everolimus trough blood levels was

    defined in either trial. A post hoc analysis of data from study

    A2306 showed that everolimus trough blood levels up to

    12 ng/mL were tolerated during 12 months of follow-up

    [34]. In line with findings from studies B201 and B251,however, in which full-dose CsA was used [26], everolimus

    Fig. 4. A significant reduction in CsA trough blood levels can be achieved

    when used in combination with everolimus. Reproduced with permission

    from Pascual [35].

  • trough blood levels between 3 and 8 ng/mL were found to

    be optimal in efficacy and safety for patients receiving

    reduced-exposure CsA.

    An unanswered question remains: how low can CNI

    blood levels be reduced in conjunction with everolimus

    without adversely influencing efficacy and safety? Compar-

    ison of the mean CsA trough blood levels of patients in

    studies B201 and A2306 showed that CsA blood levels can

    be reduced by at least 57% at 12 months when used in

    combination with everolimus (Fig. 4) [35]. Further trials

    will be needed to further study this potential for synergy.

    2.4. Calcineurin inhibitor elimination

    Calcineurin inhibitor withdrawal in everolimus-based

    regimens is currently being validated in large clinical trials.

    Studies have been conducted however with sirolimus.

    Sirolimus has been used to eliminate CsA 3 months

    posttransplant, with significantly improved graft function

    20 patients, conversion to sirolimus led to a significant

    decrease in serum creatinine from 233 F 34 to 210 F56 lmol/L after 6 months (P b .05) in 12 patients withchronic nephrotoxicity [45]. When the factors predictive of

    success in patients converted from a CNI to sirolimus were

    investigated, 27 (46%) of 59 patients were classed as

    nonresponders based on deteriorating renal function [46].

    Several baseline factors were observed that differed

    significantly between responders and nonresponders, in-

    cluding proteinuria, histological grade of allograft nephrop-

    athy, grade of vascular intimal thickening, and number of

    acute rejections before conversion. In a multivariate

    analysis, only proteinuria lower than 800 mg/d before

    conversion was a significant predictor of response [46]. This

    finding has recently been confirmed in a study of 43 patients

    converted from a CNI to sirolimus, which also identified

    baseline antihypertensive therapy and serum lactate dehy-

    drogenase level 1 month after conversion as independent

    us hav

    J. Pascual et al. / Transplantation Reviews 20 (2006) 118 7after 6, 12, 36, and 48 months in the Rapamune

    Maintenance Regimen Trial [36-41] and in other studies

    [42,43]. A meta-analysis of 6 sirolimus/CNI withdrawal

    studies, enrolling a total of 1047 patients, showed that CNI

    elimination was associated with a significant improvement

    in creatinine clearance (P b .00001) at 1 year, with asignificant reduction in hypertension (P = .0006) [44].

    There was, however, an 8% increased risk of acute rejection

    (P = .002), although the incidences of graft loss and death

    were similar in both groups. Overall, early CNI elimination

    (ie, within 23 months posttransplant) is accompanied by a

    reduction in the progression of chronic pathologic allograft

    lesions and a lower incidence of new cases and severity of

    CAN during the first year after transplantation [41]. Such

    regimens may not, however, be appropriate for patients with

    severe acute rejection episodes or persistent suboptimal

    graft function [27].

    Maintenance renal transplant patients have been con-

    verted from CsA to sirolimus. In an initial study in

    Fig. 5. Everolimus and sirolimpredictive factors [47].

    Triple therapy with sirolimus as de novo therapy has

    been compared with CsA both in combination with

    azathioprine and corticosteroids in 83 patients [48], but

    there was a high incidence of acute rejection (41% with

    sirolimus vs 38% with CsA; NS). In a trial of similar design,

    with MMF instead of azathioprine, there was a lower

    incidence of acute rejection (28% with sirolimus vs 18%

    with CsA) [49]. Two-year pooled data from these studies

    demonstrated an improvement in graft function with

    sirolimus compared with CsA (GFR, 69.3 vs 56.8 mL/min;

    P b .004) [50].Sirolimus, MMF, and corticosteroids have been used

    with basiliximab induction therapy to reduce the incidence

    of acute rejection. When this regimen was compared with

    CsA in a prospective study in 61 renal transplant recipients

    there was a numerically lower incidence of acute rejection

    (6% vs 17%) combined with significantly improved graft

    function in the sirolimus group compared with the CsA

    e similar chemical structures.

  • tic toxicity when used in combination with the CNIs or the

    3.3.1. Dyslipidemia

    Sirolimus induces dose-dependent hyperlipidemia in

    30% to 50% of patients who may require concomitant

    lipid-lowering medication [81,82]. A similar incidence of

    hypercholesterolemia and hypertriglyceridemia has been

    observed in studies of everolimus [22,25]. The alterations in

    lipid metabolism appear to be related mainly to variation in

    lipoprotein lipase activity and reduced catabolism of very

    low-density lipoprotein apolipoprotein B100 [83]. It has

    with severe refractory hyperlipidemia [33]

    Lymphocele

    ! Treat with povidone iodine and surgical intervention as required[63-65]

    Wound healing

    ! Surgical techniques to manage delayed fibrosis of wounds, for example,nonabsorbable sutures for muscle layers and delayed removal of skin

    clips [66]

    ! Avoid PSIs during wound healing in patients with diabetes orobesity [67]

    Maintenance patients

    CAN

    ! Consider reducing CsA dose to counter increased creatininelevels [33,68]

    ! Consider withdrawing CsA (if not managed by CsA reduction)[39,41,69]

    ! If CsA is reduced or withdrawn, patients may require increasedeverolimus trough blood levels [33]a

    ! If proteinuria is N800 mg/d, do not convert to PSI therapy [46]b

    Proteinuria

    ! Treat with ACE inhibitors and/or angiotensin receptor blockers [70,71]! Maintain everolimus blood trough levels at 38 ng/mL [33]! Minimization of CsA dose may be sufficient [72]Hypertension

    ! Treat with ACE inhibitors, angiotensin receptor blockers, orb-blockers [73,74]

    ! Consider withdrawal of CsA [36,43]! Use of calcium channel blockers may increase everolimus plasmaconcentrations [33]

    Anemia

    ! Treat severe anemia with erythropoietin [75]! Avoid ACE inhibitors in patients who develop anemia [70]! Reduce dose of MPA agent [76]Malignancy

    ! Use of a PSI is recommended in patients who develop a malignancyposttransplant [77,78]

    ! Consider withdrawal of CsA [38,79]a Cyclosporine withdrawal from everolimus-based regimens has not

    been validated in large clinical trials. There is limited experience of the use of

    everolimus above trough levels of 12 ng/mL [33]. A single case study reports

    everolimus trough levels above 12 ng/mL after CsAwithdrawal [80].b A phase IV study assessing 1500 mg/d proteinuria as the cutoff value

    for conversion to PSI therapy is currently enrolling patients.

    J. Pascual et al. / Transplantation Reviews 20 (2006) 1188mycophenolates, and those related to nonspecific immuno-

    suppressive actions. Management strategies for key adverse

    events are summarized in Table 3.group (creatinine clearance, 77.8 vs 64.1 mL/min at

    6 months; P = .004) [51].

    3. Proliferation signal inhibitors/mammalian targets of

    rapamycineverolimus and sirolimus

    To date, only one small head-to-head study has been

    published comparing everolimus and sirolimus in 28 patients

    [52]. In this study, renal function, measured by GFR, was

    improved using CsA in combination with everolimus rather

    than sirolimus [52]. Because of the limited analysis, a direct

    comparison of the efficacy and tolerability of the 2 agents is

    not possible. Based on review of the literature, an overview

    of these compounds is provided below.

    3.1. Pharmacology and pharmacokinetics

    Everolimus and sirolimus are macrolide derivatives

    with similar chemical structures, everolimus having a

    2-hydroxyethyl group at position 40 of the molecule

    (Fig. 5). Despite the similarities in structure, there are

    important differences in the pharmacokinetic and pharma-

    codynamic properties of the 2 molecules. Everolimus has a

    considerably shorter half-life than sirolimus (28 vs 62 hours)

    and, because of differences in regimen, everolimus reaches

    steady state in 4 days, compared with 6 days for sirolimus

    [33,53,54]. More detailed reviews of everolimus and

    sirolimus pharmacology and pharmacokinetics can be found

    in Kirchner et al [55] and Mahalati and Kahan [56].

    3.2. Interaction with CsA

    Although the modes of action of everolimus and sirolimus

    are broadly similar, there is evidence of a difference between

    these agents in their interaction with CsA. In vitro, sirolimus

    has been shown to enhance CsA activity by increasing the

    brain concentration of CsA, which in turn inhibits mito-

    chondrial glucose metabolism and high-energy phosphate

    metabolism [57,58]. Although there is no direct evidence to

    link such an effect to increased neuropathologic symptoms,

    drugs that inhibit glycolysis have the potential to enhance

    toxicity of other agents that inhibit mitochondrial oxidation

    [57]. Conversely, coadministration of everolimus and CsA

    leads to a reduction in brain CsA concentration and

    antagonizes the effect of CsA on glucose and high-energy

    phosphate metabolism [57,59]. The clinical impact of these

    different effects is not yet understood.

    3.3. Adverse events

    It has become evident, through extensive clinical studies,

    that there are specific adverse events associated with dose-

    related class actions of PSIs everolimus and sirolimus.

    Additional adverse events include those related to synergis-Table 3

    Guidelines for the management of adverse events in renal transplant

    recipients receiving everolimus as part of their immunosuppressive regimen

    De novo patients

    Hyperlipidemia

    ! Reeducate patient regarding lifestyle changes [60,61]! Treat hypercholesterolemia with statins atorvastatin, pravastatin, orfluvastatin, as preferred [25,33,61]

    ! Treat hypertriglyceridemia with fibrates [60,62]! Reevaluate the risk/benefit of continued everolimus therapy in patients

  • J. Pascual et al. / Transplantation Reviews 20 (2006) 118 9been proposed that sirolimus indirectly up-regulates expres-

    sion of the gene apo CIII, an important inhibitor of

    lipoprotein lipase [27,84]. Sirolimus has also been shown

    to lead to a 42% increase in free fatty acid concentrations,

    thus stimulating hepatic triglyceride synthesis [85].

    Hyperlipidemia should be managed in accordance with

    guidelines (eg, National Kidney Foundation Kidney Disease

    Outcomes Quality Initiative), focusing on both lifestyle

    changes and drug treatment [60,61]. Statins and fibrates are

    used to treat elevated low-density lipoprotein cholesterol

    and hypertriglyceridemia, respectively [60]. Lipid-lowering

    agents (mainly statins) were used in 58.9% and 66.4% of

    patients receiving everolimus 1.5 and 3.0 mg/d in study

    A2306, whereas in study A2307, 66.7% and 72.7% of

    patients received such therapy [25]. In addition, statins were

    taken by 90% of patients after heart transplantation;

    although low-density lipoprotein and high-density lipopro-

    tein levels were increased at 12 months, there was no

    difference between the azathioprine and everolimus groups

    [86]. In a study of healthy individuals given single doses of

    everolimus in addition to atorvastatin or pravastatin, no

    clinically significant effects on the pharmacokinetics of any

    drug were noted [87], although fluvastatin has recently been

    shown to improve cardiovascular outcomes in renal

    transplant recipients in a large-scale clinical trial [88].

    3.3.2. Myelosuppression

    A higher incidence of microcytic anemia, perhaps related

    to antiproliferative effects in bone marrow and reduced

    incorporation of iron, has been observed with sirolimus than

    with CsA or MMF in a number of studies [81,89,90]. In a

    12-month study comparing everolimus and MMF (study

    B201), the incidence of anemia was similar with both

    agents: 34.3% in patients receiving everolimus 3.0 mg/d vs

    32.1% in those receiving MMF [22].

    Thrombocytopenia is common with PSIs, but seldom

    clinically significant. In phase III trials with everolimus and

    low-dose CsA, the incidence was 3.6% and 8.0% for

    everolimus 1.5 and 3.0 mg/d, respectively, in the absence of

    IL-2 receptor agonist therapy, and 3.4% and 5.8% with IL-2

    receptor therapy [25]. Thrombocytopenia occurred in 37%

    to 45% of sirolimus and 9% to 23% of sirolimus/CsA-

    treated recipients [48,49,81,82]. The incidence, but not the

    severity, of thrombocytopenia correlated with sirolimus

    trough blood levels; however, this is self-limiting and the

    adverse event disappears over time [27]. The incidence of

    leukopenia in everolimus studies is similar to that of

    thrombocytopenia, occurring in around 4% of patients in

    the absence of IL-2 receptor agonist therapy, and in 3.4%

    (1.5 mg/d dose) and 5.8% (3.0 mg/d) with IL-2 receptor

    therapy [25].

    Two hypotheses have been suggested to explain the

    myelosuppression observed with PSIs. First, in vitro studies

    have shown enhanced, dose-dependent, agonist-induced

    platelet aggregation and granule secretion after sirolimus

    exposure. Second, sirolimus inhibits signal transduction viathe gp130b chain, which is shared by cytokine receptors for

    IL-11, granulocyte colony-stimulating factor, and erythro-

    poietin. These molecules are all necessary for the production

    of platelets and leukocytes, and sirolimus may therefore

    inhibit their production [27].

    Dose reduction of PSIs may be an appropriate response

    to myelosuppression, but evidence for this is limited, and

    active treatment with erythropoietin may become necessary

    in severe anemia [90].

    3.3.3. Edema

    Edema of the lower and upper limbs is commonly

    observed during treatment with sirolimus or everolimus

    [27,91]. Prostacyclin release from endothelial cells, stimu-

    lated by sirolimus, leads to vasodilatation [92], suggesting

    that edema may be related to capillary leakage, as has been

    observed in sirolimus-treated lung transplant recipients and

    patients with psoriasis [93,94]. In addition to limb edema,

    bilateral eyelid edema has been observed both in de novo

    and maintenance transplant recipients receiving sirolimus

    and everolimus [91,95,96].

    3.3.4. Arthralgia

    Arthralgia is a common adverse event occurring with

    both drugs. In a study comparing sirolimus and CsA,

    arthralgia was reported in 20% of patients in the sirolimus

    group at 12 months, but was not reported in the CsA group

    [48]. In a separate study comparing sirolimus and azathio-

    prine, arthralgia was listed as a cause of study discontinu-

    ation in the sirolimus group [81]. Arthralgia has also been

    reported in patients in the everolimus A2306 study [91]. The

    cause of this phenomenon is, however, not clear.

    3.3.5. Impaired wound healing

    The antiproliferative effects of PSIs have been associated

    with a high incidence of wound healing problems. Delayed

    healing of lymphatic channels divided during surgery and

    a reduced fibrotic reaction contribute to an increased

    incidence of lymphocele [97]. Compared with azathioprine,

    sirolimus 5 mg/d is associated with an increased incidence

    of postoperative lymphoceles [81], and similar results have

    been seen with sirolimus in combination with CsA or

    tacrolimus [98]. In studies A2306 and A2307, the incidence

    of lymphocele at 6 months posttransplant was 15.2% and

    10.3% with everolimus 1.5 mg/d, and 6.4% and 7.2% with

    everolimus 3.0 mg/d, respectively [25] (Table 4). Potential

    risk factors for delayed healing include recipient age,

    presence of diabetes, and living vs cadaveric donor

    [66,67]. In addition, thymoglobulin induction, body mass

    index greater than 32 kg/m2, and a cumulative sirolimus

    dose greater than 35 mg over the first 5 days of treatment

    were all significantly associated with impaired wound

    healing [98]. When the loading dose of sirolimus was

    eliminated from a study of steroid-free maintenance

    regimens in 239 patients, the incidence of wound healing

    problems decreased [99]. Excellent surgical technique, such

  • hibiting renal tubular cell regeneration and by increasing

    enal tubular cell loss by apoptosis [103].

    .3.7. Proteinuria

    In chronic glomerular diseases, proteinuria is an impor-

    nt marker of the risk of a progressive, irreversible decline

    GFR [104], and proteinuria levels greater than 800 mg/d

    ave a negative correlation with outcome if treatment of

    atients is converted from CNI to sirolimus [46]. Minimi-

    ation of proteinuria is, therefore, a key goal in the treatment

    f chronic kidney disease [70]. In 50 renal transplant

    ecipients with CAN who were switched from CNIs to

    irolimus, 64% developed marked proteinuria, with ne-

    hrotic syndrome in around half of these patients [105],

    atients with preexisting proteinuria having a higher

    ecurrence rate. In 1 study of patients with existing CAN,

    2% had no proteinuria (b150 mg/d) at the time ofonversion from CNI to sirolimus [47]. However, 19.4%

    f patients had proteinuria greater than 1000 mg/d at 1-year

    inhibitor regimens are safe and effective in the long-term

    treatment of hypertension in renal transplant recipients

    [73,74]. The long-term benefits of using calcium channel

    blockers are not well defined in renal transplant recipients,

    and there may be a possible interaction between agents

    such as verapamil or diltiazem with everolimus, resulting in

    an increase in everolimus levels [33]. Studies with

    sirolimus have shown a significant reduction in blood

    pressure after CNI elimination, demonstrating another

    possible approach [36,43].

    4. Proliferation signal inhibitors in current

    clinical practice

    4.1. Patient selection

    In combination with CsA and corticosteroids, everolimus

    is currently approved for the prophylaxis of organ rejection

    in adult patients at low-to-moderate immunologic risk

    s and

    /d)a

    /d)a

    /d)b

    /d)b

    .0 mg

    .0 mg/d) 154

    mus

    blood

    blood

    /d)

    /d)

    J. Pascual et al. / Transplantation Reviews 20 (2006) 11810r

    in

    r

    3

    ta

    in

    h

    p

    z

    o

    r

    s

    p

    p

    r

    8

    c

    oas meticulous ligation of transected lymphatic channels,

    delayed removal of skin clips, and the use of nonabsorbable

    sutures, may reduce wound complications. Management of

    posttransplant lymphocele remains the subject of debate.

    Milder cases may be self-limiting or respond to instillation

    of povidone iodine [63], although laparoscopic drainage

    may be required [64,65].

    3.3.6. Renal dysfunction

    Experimental data demonstrate that the PSI class of

    drugs has limited direct toxic effects on the kidney;

    however, sirolimus and everolimus clearly augment CNI

    toxicity partly through the effect shown with sirolimus,

    which increases tissue CsA concentrations [100,101]

    and may also be related to its effects on glucose meta-

    bolism [102].

    There has been concern that PSIs delay the recovery of

    enal function after acute tubular necrosis (ATN) possibly by

    Table 4

    Incidence of wound healing problems and lymphoceles in trials of sirolimu

    Study Duration (mo) Agent

    Study A2306 [25] 6 Everolimus (1.5 mg

    Everolimus (3.0 mg

    Study A2307 [25] 6 Everolimus (1.5 mg

    Everolimus (3.0 mg

    Kahan [81] 12 CsA + sirolimus (2

    CsA + sirolimus (5

    CsA + azathioprine

    Ciancio et al [98] 12 Tacrolimus + siroli

    Tacrolimus + MMF

    CsA + sirolimus

    Studies B201 and B251 12 Everolimus (trough

    Everolimus (trough

    Study B201 [23] 36 Everolimus (1.5 mg

    Everolimus (3.0 mg

    MMF (2.0 g/d)

    a In combination with reduced-dose CsA.b In combination with reduced-dose CsA and basiliximab induction.

    4 P b .001 vs azathioprine.postconversion and this increased to 20.6% at 2 years [47].

    Experimental laboratory evidence suggests that sirolimus

    may lead to an increase in proteinuria through interference

    with normal tubular epithelial cell compensatory mecha-

    nisms [106]. It is of interest that there was no excess de novo

    proteinuria reported in any of the randomized phase II and

    III studies of sirolimus [27]. There is no evidence available

    for everolimus in phase III studies as data on proteinuria

    were not routinely collected. However, the incidence,

    mechanism, and management of proteinuria in patients

    receiving everolimus are currently being investigated.

    Angiotensin-converting enzyme (ACE) inhibitors and

    angiotensin II receptor blockers may have potential clinical

    utility for the management of both hypertension and

    proteinuria in patients receiving everolimus [71,104]. This

    is an area that requires further investigation especially in

    relation to exacerbating anemia [107].

    Nevertheless, clinical trial data suggest that ACE

    3

    16 6

    6 4

    14 4

    level, 3 ng/mL) 20 20

    level, 8 ng/mL) 12 16

    9

    12

    4 everolimus

    Lymphocele (%) Wound dehiscence (%)

    15.2

    6.4

    10.3

    7.2

    /d) 12

  • J. Pascual et al. / Transplantation Reviews 20 (2006) 118 11receiving an allogeneic renal or cardiac transplant. Ever-

    olimus should not be used with full doses of CsA beyond

    1 month posttransplantation.

    4.1.1. bOld-for-oldQ transplantationIn recent years, the age of patients receiving renal

    transplants has increased, with around one third being older

    than 55 years [108,109]. Similarly, the proportion of donors

    aged older than 55 years has increased. There has therefore

    been a trend toward age matching of donor and recipient to

    ensure appropriate use of donated organs [80,108,110,111].

    This means that the current estimated potential for bold-for-old Q transplantation is approximately 30% of all trans-plants. Older organs are at greater risk of delayed graft

    functioning, CAN and CNI toxicity, and it has been

    suggested that they should be targeted for CNI minimization

    [80,110,112].

    4.1.2. Patients younger than 15 years

    Everolimus is not currently indicated in pediatric

    patients, but dosing, efficacy, and safety data in this patient

    group are beginning to emerge [113,114].

    4.1.3. Malignancy

    Everolimus and sirolimus interrupt the PI3K/Akt signal-

    ing pathway, which plays a critical role in regulating cell

    proliferation, survival, mobility, and angiogenesis [115].

    Tumors that rely on overactivity of the PI3K/Akt pathway

    for growth may therefore be susceptible to this class of

    agent. In vitro and in vivo studies have shown that growth

    of a number of tumor cell lines can be slowed or prevented

    with sirolimus, including those derived from liver [116,117],

    breast [118], pancreas [119], and ovary [120], as well as

    myeloid and lymphocytic leukemia cell lines [121,122]. In a

    breast cancer cell line, sirolimus has also been shown to

    restore sensitivity to tamoxifen [123].

    Use of everolimus in vitro and in vivo has yielded

    similar results to those with sirolimus. A combination of

    everolimus and epidermal growth factor receptor/vascular

    endothelial growth factor receptor 2 leads to inhibition

    of glioblastoma growth [124], and everolimus has also

    been shown to reverse Akt-dependent prostate intra-

    epithelial neoplasia [125]. In a primary cell culture derived

    from patients with acute myeloid leukemia, everolimus

    enhanced the activity of the PI3 kinase inhibitor,

    LY294002 [126]. Everolimus has also been shown to

    inhibit the growth of transformed B lymphocytes [127] and

    a cell line derived from a patient with posttransplant

    lymphoproliferative disorder [128]. In a rat pancreatic

    tumor model, everolimus demonstrated significant, dose-

    dependent antitumor activity that was similar to that

    observed with the cytotoxic agent 5-fluorouracil [129]. In

    a study combining everolimus with the DNA-damaging

    agent cisplatin, everolimus was shown to inhibit expres-

    sion of p21, thus increasing the susceptibility of tumor

    cells to apoptosis [130].In phase III studies of sirolimus, no increased incidence

    of malignancies has been observed compared with either

    placebo or azathioprine when combined with CsA and

    corticosteroids [81,82]. During a 3-year study of CNI

    elimination in sirolimus-treated patients, those in whom

    CNI therapy was stopped had a numerically lower incidence

    of malignancies compared with those remaining on CNI

    (5.6% vs 11.2%) [37]. When results were pooled from

    5 phase II and III studies, the overall incidence of

    malignancies in patients receiving a combination of CsA

    and sirolimus was found to be similar to that in patients

    receiving CsA or azathioprine, whereas early elimination of

    CsA was associated with a reduced risk of malignancies

    [79]. In a multicenter, open-label study of 167 patients

    enrolled from previous sirolimus studies (mean exposure to

    sirolimus, 1526 days), malignancies were reported in 11

    patients receiving CsA and sirolimus; none of the patients in

    whom CsA had been eliminated experienced malignancies

    [38]. In a study of 430 patients randomized at 3 months after

    renal transplantation to continue sirolimus and CsA or have

    CsA withdrawn, CNI-free therapy significantly reduced the

    incidence of nonskin cancer at 5 years posttransplant [131].

    In studies of everolimus, the incidence of malignancies was

    lowafter 36 months of treatment, the incidence of

    malignancies was 5.2%, 4.5%, and 4.6% in the 1.5 mg/d

    everolimus, 3.0 mg/d everolimus, and MMF group, respec-

    tively (study B201) [23], and 4.7%, 5.2%, and 6.1%,

    respectively (study B251) [24]. After 24 months of

    treatment with reduced CsA doses, the proportion of

    patients experiencing malignancies was even lower [30].

    Conversion from CsA to sirolimus has recently been

    suggested as a potential method of treating malignancies in

    transplant recipients without increasing the risk of graft

    rejection. For example, 2 renal transplant recipients with

    Kaposi sarcoma underwent conversion from CsA to siroli-

    mus, with complete regression of their Kaposi sarcoma

    lesions [77]. These beneficial effects on Kaposi sarcoma

    were further explored in a prospective study in 15 renal

    transplant recipients who were switched from CsA to

    sirolimus [78]. Three months after conversion, all Kaposi

    sarcoma lesions had disappeared, and remission was con-

    firmed by histopathology at 6 months. There were no

    acute episodes of rejection or changes in graft function.

    Beneficial effects of sirolimus in liver transplant recipients

    with hepatocellular carcinoma have also been reported,

    in both decreased tumor recurrence and remission of

    metastases [132,133].

    4.2. Everolimus dosing and administration

    As has been demonstrated in clinical trials, everolimus is

    an effective immunosuppressant when given immediately

    after renal transplantation in combination with CsA,

    corticosteroids and, possibly, IL-2 receptor antagonist

    induction therapy [21,25]. Suggested algorithms for ever-

    olimus treatment are provided for de novo (Fig. 6) and

    maintenance (Fig. 7) renal transplant recipients.

  • J. Pascual et al. / Transplantation Reviews 20 (2006) 118124.2.1. De novo patients

    In de novo renal transplant recipients, everolimus should

    be initiated at a dose of 0.75 mg BID dose adjustment to

    ensure trough blood levels of 3 to 8 ng/mL [26,34] (Fig. 6).

    Straightforward TDM can now be achieved using an

    immunoassay (Innofluor Certican, Seradyn Inc, Indian-

    apolis, Ind). Lower CsA C2 levels can be targeted in the

    weeks after transplantation with IL-2 receptor antibody

    Fig. 6. Treatment guidelines for the use of everolimus in de novo renal transplan

    stable at with down-titration of CsA concentrations, with CsA C0 levels in t

    therapy, CsA exposure may be minimized further: CsA C2 levels 500 to 700

    [25,33]. yCsA withdrawal from everolimus-based regimens is currently being veverolimus above trough levels of 12 ng/mL [33]. A single case study reports e

    indicates twice daily.induction [25]. Table 5 provides insights from a single

    center experience on the practical use of low-dose CsA and

    everolimus in renal transplantation.

    4.2.2. Maintenance patients

    Based on studies with sirolimus, everolimus could allow

    CsA minimization or elimination [38,44,46] (Fig. 7).

    Cyclosporine has been reduced or withdrawn in progres-

    t recipients. Everolimus trough blood levels have been reported to remain

    he range of 25 to 50 ng/mL [26,134,135]. *With basiliximab induction

    ng/mL in weeks 0 to 8 and 350 to 450 ng/mL in week 9 to month 12

    alidated in large clinical trials. zThere is limited experience of the use ofverolimus trough levels above 12 ng/mL after CsA withdrawal [80]. b.i.d.

  • Fig. 7. Treatment guidelines for the use of everolimus in maintenance renal transplant recipients receiving CNIs. CNI can be reduced in a stepwise progression

    of approximately 25% each step; however, abrupt cessation of CNI is also used in clinical practice. zRange 800 to 1500 mg/d; published data from 1 study [46]recommends 800 mg/d as the cutoff point for proteinuria; however, an ongoing phase IV study is assessing 1500 mg/d proteinuria as the cutoff value for

    conversion to PSI therapy. yThere is limited experience of the use of everolimus above trough levels of 12 ng/mL [33]. Clinical opinion (see Table 5) suggeststhat lower everolimus exposure may allow for increased tolerability. Everolimus levels of 6 to 12 ng/mL are currently being evaluated in ongoing phase IV

    studies. *CsA withdrawal from everolimus-based regimens is currently being validated in large clinical trials.

    J. Pascual et al. / Transplantation Reviews 20 (2006) 118 13

  • insig

    [80] p

    eg, y

    of thi

    toxic

    lder p

    e to CNI-related nephrotoxicity, not least because older organs tend to have worse

    renal

    re to

    case

    to 40

    sA. G

    splant

    ld-for

    from

    us an

    l tran

    receiv

    J. Pascual et al. / Transplantation Reviews 20 (2006) 11814Table 5

    Low-dose CsA and everolimus strategies in de novo renal transplantation

    Q: Which renal transplant patients should be considered for PSI therapy?

    A: Everolimus offers potential for both CsA minimization and withdrawal

    ! De novo patients at high risk of suboptimal renal function! Patients for whom very long-term immunosuppression will be required (! Patients at risk for CMV infection (it is associated with a low incidence! In the maintenance setting, patients at risk for or exhibiting CNI-related

    Q: Can one define patients at high risk of suboptimal graft function?

    A: In an effort to expand the donor pool, it is becoming common for o

    transplantation [80,137]. However, these patients may be more susceptibl

    function than younger organs [112,136].

    Q: How may everolimus be used to reduce CNI exposure and to optimize

    A: Two recent clinical trials show that everolimus allows CsA exposu

    immunosuppressive efficacy and good renal function [25]. Two long-term

    recently published [80].

    ! Case 1: Everolimus 0.75 mg BID allowed a reduction in CsA C2 levelsstabilized at around 1.7 mg/dL [80].

    ! Case 2: Everolimus 1.5 mg BID was combined with reduced-exposure Cpeaking at 3.8 mg/dL before CsAwas withdrawn [80]. By 2 years posttran

    everolimus and low-dose steroids [80].

    Q: What is an acceptable posttransplant mean serum creatinine level for boA: Previous experience from Spain shows that patients receiving a kidney

    2.06 mg/dL [112]. Case 1 above, in which a patient treated with everolim

    2 years posttransplant represents a good outcome for an old-for-old rena

    Q: What CsA C2 levels optimize long-term renal graft function in patientssive steps of approximately 25% over 4 weeks; how-

    ever, immediate cessation of CNIs has also been used in

    clinical practice.

    In the maintenance phase of treatment, there are currently

    few data in patients with CsAC2 levels lower than 350 ng/mL

    (trough blood (C0) 50 ng/mL). Initial experience suggests

    that CsA can be discontinued in selected patients receiving

    everolimus [80], although the dose of everolimus then needs

    to be increased to compensate for the pharmacokinetic

    interaction that leads to a 2- to 3-fold decrease in everolimus

    blood levels when CsA is withdrawn [15]. After CsA

    withdrawal, initial clinical experience suggests that ever-

    olimus trough blood levels of 10 to 15 ng/mL may be needed

    when using everolimus and corticosteroids only [80], but that

    lower trough blood levels may be required if also using an

    MPA agent.

    4.1.3. Future study of everolimus

    There is an ongoing phase IV clinical trial program for

    everolimus to refine the use of everolimus in clinical

    practice [7], including a delayed start to permit wound

    healing, prospective use in maintenance renal transplant

    patients, and the ability to facilitate CNI minimization and

    withdrawal. Everolimus is currently licensed for use in

    combination with low-dose CsA, but tacrolimus is also

    widely used in renal transplant recipients. At least 1

    pharmacokinetic phase II and 2 large phase III trials are

    exploring the combination of tacrolimus and everolimus in

    de novo kidney transplantation. Everolimus has also been

    shown to reduce the severity and incidence of cardiac

    allograft vasculopathy posttransplantation in heart transplan

    recipients, perhaps by inhibiting proliferation of smooth

    muscle cells [86]. This is an additional area for future

    investigation in the renal transplant population.

    5. Conclusions

    There is good clinical trial evidence to support the

    efficacy and tolerability of everolimus in renal transplanta-

    tion. Clinical trial data indicate that everolimus can facilitate

    minimization of CsA exposure, but ongoing clinical trials

    and clinical practice will help to further refine its therapeutic

    role. Everolimus is associated with several class-specific

    adverse events (eg, hyperlipidemia), but experience to date

    A: Minimization of CsA exposure to ensure C2 levels less than 400 ng/mL by 6 months posttransplant is desirable [80]. Cyclosporine C2 exposure levels o

    350450 ng/mL from week 13 posttransplantation, or week 9 posttransplantation if using basiliximab (Simulect) induction therapy, are recommended

    [25,33].

    Q: When is complete withdrawal of CNIs appropriate?

    A: CNI withdrawal is generally only necessary if the patient is experiencing nephrotoxicity or other serious adverse events that do not respond to CNI dose

    minimization [80]. In our experience, if CNIs are withdrawn, everolimus dose should be increased to achieve blood trough levels of 812 ng/mL, but there

    are no clinical trials to support this strategy.

    Q: Which specific adverse events associated with use of everolimus are most troublesome?

    A: Patients may experience hyperlipidemia during everolimus therapy, which can generally be controlledwith statins [33,80]. Early after transplantation, lymphocele

    or wound dehiscence can occur, but they are usually not severe [33]. Lymphocele may be self-limiting or responsive to povidone iodine treatment [91].

    Q: How should everolimus be used in combination with mycophenolate therapy in maintenance renal transplant recipients?

    A: It is advisable to closely monitor patients receiving everolimus, MPA, and steroids to ensure that they do not become over-immunosuppressed or anemic

    [138]. The use of low levels of everolimus (38 ng/mL) and MMF (1 g) should be considered. Formal study in clinical trials is needed to explore this strategyt

    f

    .function?

    be reduced in de novo renal transplant recipients, while maintaining

    studies of bold-for-oldQ renal transplants from one of these trials have been

    0 ng/mL after 4 months posttransplant [80]. Serum creatinine subsequently

    raft function deteriorated despite CsA minimization, with serum creatinine

    , serum creatinine had stabilized at 2.5 mg/dL with the patient maintained on

    -oldQ transplant patients?an elderly donor have mean 12-month serum creatinine levels around

    d reduced-dose CsA has a stable serum creatinine level of 1.7 mg/dL up to

    splant [80].

    ing everolimus?hts from Hospital Ramon y Cajal, Madrid, Spain

    articularly in:

    oung patients) [136]

    s infection [22])

    ities or CAN.

    atients (ie, N55 years) to receive older kidneysso-called bold-for-oldQ

  • microemulsion in cynomolgus monkey kidney allotransplantation.

    the novel rapamycin analog, SDZ RAD, to rat lung allograft

    recipients: potentiation of immunosuppressive efficacy and improve-

    J. Pascual et al. / Transplantation Reviews 20 (2006) 118 15ment of tolerability of staggered versus simultaneous treatment.

    Transplantation 1999;67:956 -62.

    [13] Lutz J, Zou H, Liu S, Antus B, et al. Apoptosis and treatment of

    chronic allograft nephropathy with everolimus. Transplantation

    2003;76:508 -15.

    [14] Neumayer HH, Paradis K, Korn A, et al. Entry-into-human study

    with the novel immunosuppressant SDZ RAD in stable renal

    transplant recipients. Br J Clin Pharmacol 1999;48:694 -703.

    [15] Kovarik JM, Kalbag J, Figueiredo J, et al. Differential influence of

    two cyclosporine formulations on everolimus pharmacokinetics: a

    clinically relevant pharmacokinetic interaction. J Clin Pharmacol

    2002;42:95 -9.Transplantation 2000;69:737 -42.

    [11] Viklicky O, Zou H, Muller V, et al. SDZ-RAD prevents manifes-

    tation of chronic rejection in rat renal allografts. Transplantation

    2000;69:497-502.

    [12] Hausen B, Boeke K, Berry GJ, et al. Coadministration of Neoral andsuggests that these can be managed. Investigation of

    the antineoplastic activity of everolimus is anticipated given

    the positive effects that have been documented for sirolimus

    especially in patients with Kaposi sarcoma. This class

    of agent has yet to find its true role, but the potential

    to maintain immunosuppression without the twin penalties

    of nephrotoxicity and malignancy will provide the

    incentive to refine our clinical use of both sirolimus

    and everolimus.

    Acknowledgement

    The authors thank Dr J Chapman for review of the

    manuscript.

    References

    [1] UNOS. United Network for Organ Sharing. http://www.optn.org/

    AR2004/111a_dh.htm [Last accessed: June 2005].

    [2] Racusen LC, Solez K, Colvin RB, et al. The Banff 97 working clas-

    sification of renal allograft pathology. Kidney Int 1999;55:713-23.

    [3] Nashan B. The role of Certican (everolimus, RAD) in the many

    pathways of chronic rejection. Transplant Proc 2001;33:3215-20.

    [4] Nankivell BJ, Borrows RJ, Fung CL, et al. The natural history of

    chronic allograft nephropathy. N Engl J Med 2003;349:2326 -33.

    [5] Schuler W, Sedrani R, Cottens S, et al. SDZ RAD, a new rapamycin

    derivative: pharmacological properties in vitro and in vivo.

    Transplantation 1997;64:36 -42.

    [6] Schuurman HJ, Pally C, Weckbecker G, et al. C. SDZ RAD inhibits

    cold ischemia-induced vascular remodeling. Transplant Proc

    1999;31:1024-5.

    [7] Neumayer HH. Introducing everolimus (Certican) in organ trans-

    plantation: an overview of preclinical and early clinical develop-

    ments. Transplantation 2005;79:S72-5.

    [8] Schuurman HJ, Cottens S, Fuchs S, et al. SDZ RAD, a new

    rapamycin derivative: synergism with cyclosporine. Transplantation

    1997;64:32-5.

    [9] Schuurman HJ, Schuler W, Ringers J, et al. The macrolide SDZ RAD

    is efficacious in a nonhuman primate model of allotransplantation.

    Transplant Proc 1998;30:2198-9.

    [10] Schuurman HJ, Ringers J, Schuler W, et al. Oral efficacy of the

    macrolide immunosuppressant SDZ RAD and of cyclosporine[16] Kovarik JM, Dantal J, Civati G, et al. Influence of delayed initiation

    of cyclosporine on everolimus pharmacokinetics in de novo renal

    transplant patients. Am J Transplant 2003;3:1576 -80.

    [17] Kirchner GI, Winkler M, Mueller L, et al. Pharmacokinetics of SDZ

    RAD and cyclosporin including their metabolites in seven kidney

    graft patients after the first dose of SDZ RAD. Br J Clin Pharmacol

    2000;50:449-54.

    [18] Kahan BD, Wong RL, Carter C, et al. A phase I study of a 4-week

    course of SDZ-RAD (RAD) in quiescent cyclosporine-prednisone

    treated renal transplant recipients. Transplantation 1999;68:1100-6.

    [19] Kovarik JM, Kahan BD, Kaplan B, et al. Longitudinal assessment of

    everolimus in de novo renal transplant recipients over the first post-

    transplant year: pharmacokinetics, exposure-response relationships,

    and influence on cyclosporine. Clin Pharmacol Ther 2001;69:48-56.

    [20] Kahan BD, Kaplan B, Lorber MI, et al. RAD in de novo renal

    transplantation: comparison of three doses on the incidence and

    severity of acute rejection. Transplantation 2001;71:1400-6.

    [21] Nashan B, Curtis J, Ponticelli C, et al. Everolimus and reduced-

    exposure cyclosporine in de novo renal-transplant recipients: a three-

    year phase II, randomized, multicenter, open-label study. Transplan-

    tation 2004;78:1332-40.

    [22] Vtko S, Margreiter R, Weimar W, et al. Everolimus (Certican)

    12-month safety and efficacy versus mycophenolate mofetil in de

    novo renal transplant recipients. Transplantation 2004;78:1532-40.

    [23] Vtko S, Margreiter R, Weimar W, et al. Three-year efficacy and

    safety results from a study of everolimus versus mycophenolate

    mofetil in de novo renal transplant patients. Am J Transplant

    2005;5:2521-30.

    [24] Lorber MI, Mulgaonkar S, Butt KMH, et al. Everolimus versus

    mycophenolate mofetil in the prevention of rejection in de novo renal

    transplant recipients: a 3-year randomized, multicenter, phase III

    study. Transplantation 2005;80:244-52.

    [25] Vtko S, Tedesco H, Eris J, et al. Everolimus with optimized

    cyclosporine dosing in renal transplant recipients: 6-month safety

    and efficacy results of two randomized studies. Am J Transplant

    2004;4:626-35.

    [26] Lorber MI, Ponticelli C, Whelchel J, et al. Therapeutic drug

    monitoring for everolimus in kidney transplantation using 12-month

    exposure, efficacy, and safety data. Clin Transplant 2005;19:145 -52.

    [27] Kuypers DR. Benefit-risk assessment of sirolimus in renal trans-

    plantation. Drug Saf 2005;28:153 -81.

    [28] Holmes M, Chilcott J, Walters S, et al. Economic evaluation of

    everolimus versus mycophenolate mofetil in combination with

    cyclosporine and prednisolone in de novo renal transplant recipients.

    Transpl Int 2004;17:182-7.

    [29] Leone J, Vtko S, Whelchel J, et al. Excellent graft function in de

    novo kidney transplant recipients treated with Certican, Simulect and

    reduced Neoral exposure: 24 month results. Am J Transplant

    2005;5(Suppl 11):A1010.

    [30] Pascual J, Cambi V, Dissegna D, et al. Efficacy and safety of 2 doses

    of everolimus combined with reduced dose Neoral in de novo kidney

    transplant recipients: 24 months analysis. Am J Transplant

    2005;5(Suppl 11):A1010.

    [31] Magee J, Tedesco H, Pascual J, et al. Efficacy and safety of 2 doses

    of everolimus combined with reduced-dose Neoral in de novo kidney

    transplant recipients: 12 month analysis. Am J Transplant 2004;

    4(Suppl 8):296 [abstract 504].

    [32] Whelchel J, Vitko S, Eris J, et al. Excellent graft function in de novo

    kidney transplant recipients treated with Certican, Simulect and

    reduced Neoral exposure. 12-month results. Am J Transplant

    2004;4(Suppl 8):297 [abstract 507].

    [33] Novartis Pharma AG. Certican summary of product characteristics.

    Basel7 Novartis Pharma AG; 2003.

    [34] Kovarik JM, Tedesco H, Pascual J, et al. Everolimus therapeutic

    concentration range defined from a prospective trial with reduced-

    exposure cyclosporine in de novo kidney transplantation. Ther Drug

    Monit 2004;26:499-505.

  • J. Pascual et al. / Transplantation Reviews 20 (2006) 11816[35] Pascual J. Concentration-controlled everolimus (Certican): combi-

    nation with reduced dose calcineurin inhibitors. Transplantation

    2005;79:S76 -9.

    [36] Johnson RWG, Kreis H, Oberbauer R, et al. Sirolimus allows

    early cyclosporine withdrawal in renal transplantation resulting in

    improved renal function and lower blood pressure. Transplantation

    2001;72:777 -86.

    [37] Kreis H, Oberbauer R, Campistol JM, et al. Long-term benefits with

    sirolimus-based therapy after early cyclosporine withdrawal. J Am

    Soc Nephrol 2004;15:809 -17.

    [38] Morales JM, Campistol JM, Kreis H, et al. Sirolimus-based therapy

    with or without cyclosporine: long-term follow-up in renal transplant

    patients. Transplant Proc 2005;37:693-6.

    [39] Mota A, Arias M, Taskinen EI, et al. Sirolimus-based therapy

    following early cyclosporine withdrawal provides significantly

    improved renal histology and function at 3 years. Am J Transplant

    2004;4:953 -61.

    [40] Oberbauer R, Segoloni G, Campistol JM, et al. Early cyclosporine

    withdrawal from a sirolimus-based regimen results in better renal

    allograft survival and renal function at 48 months after transplanta-

    tion. Transpl Int 2005;18:22 -8.

    [41] Ruiz JC, Campistol JM, Grinyo JM, et al. Early cyclosporine a

    withdrawal in kidney-transplant recipients receiving sirolimus

    prevents progression of chronic pathologic allograft lesions. Trans-

    plantation 2004;78:1312-8.

    [42] Baboolal K. A phase III prospective, randomized study to evaluate

    concentration-controlled sirolimus (rapamune) with cyclosporine

    dose minimization or elimination at 6 months in de novo renal

    allograft recipients. Transplantation 2003;75:1404-8.

    [43] Gonwa TA, Hricik DE, Brinker K, et al. Improved renal function in

    sirolimus-treated renal transplant patients after early cyclosporine

    elimination. Transplantation 2002;74:1560-7.

    [44] Mulay AV, Hussain N, Fergusson D, et al. Calcineurin inhibitor

    withdrawal from sirolimus-based therapy in kidney transplantation: a

    systematic review of randomized trials. Am J Transplant 2005;5:

    1748-56.

    [45] Dominguez J, Mahalati K, Kiberd B, et al. Conversion to rapamycin

    immunosuppression in renal transplant recipients: report of an initial

    experience. Transplantation 2000;70:1244-7.

    [46] Diekmann F, Budde K, Oppenheimer F, et al. Predictors of success in

    conversion from calcineurin inhibitor to sirolimus in chronic

    allograft dysfunction. Am J Transplant 2004;4:1869-75.

    [47] Bumbea V, Kamar N, Ribes D, et al. Long-term results in renal

    transplant patients with allograft dysfunction after switching from

    calcineurin inhibitors to sirolimus. Nephrol Dial Transplant 2005;

    20:2517-23.

    [48] Groth CG, B7ckman L, Morales JM, et al. Sirolimus (rapamycin)based therapy in human renal transplantation: similar efficacy and

    different toxicity compared with cyclosporine. Sirolimus European

    Renal Transplant Study Group. Transplantation 1999;67:1036-42.

    [49] Kreis H, Cisterne JM, Land W, et al. Sirolimus in association with

    mycophenolate mofetil induction for the prevention of acute graft

    rejection in renal allograft recipients. Transplantation 2000;69:

    1252-60.

    [50] Morales JM, Wramner L, Kreis H, et al. Sirolimus does not exhibit

    nephrotoxicity compared to cyclosporine in renal transplant recipi-

    ents. Am J Transplant 2002;2:436 -42.

    [51] Flechner SM, Goldfarb D, Modlin C, et al. Kidney transplantation

    without calcineurin inhibitor drugs: a prospective, randomized trial

    of sirolimus versus cyclosporine. Transplantation 2002;74:1070-6.

    [52] Kamar N, Allard J, Ribes D, et al. Assessment of glomerular and

    tubular functions in renal transplant patients receiving cyclosporine

    A in combination with either sirolimus or everolimus. Clin Nephrol

    2005;63:80 -6.

    [53] Ferron GM, Mishina EV, Zimmerman JJ, et al. Population

    pharmacokinetics of sirolimus in kidney transplant patients. Clin

    Pharmacol Ther 1997;61:416-28.[54] MacDonald A, Scarola J, Burke JT, et al. Clinical pharmacokinetics

    and therapeutic drug monitoring of sirolimus. Clin Ther 2000;

    22(Suppl B):B101-21.

    [55] Kirchner GI, Meier-Wiedenbach I, Manns MP. Clinical pharmaco-

    kinetics of everolimus. Clin Pharmacokinet 2004;43:83-95.

    [56] Mahalati K, Kahan BD. Clinical pharmacokinetics of sirolimus. Clin

    Pharmacokinet 2001;40:573-85.

    [57] Serkova N, Jacobsen W, Niemann CU, et al. Sirolimus, but not the

    structurally related RAD (everolimus), enhances the negative effects

    of cyclosporine on mitochondrial metabolism in the rat brain. Br J

    Pharmacol 2001;133:875-85.

    [58] Serkova N, Litt L, James TL, et al. Evaluation of individual and

    combined neurotoxicity of the immunosuppressants cyclosporine

    and sirolimus by in vitro multinuclear NMR spectroscopy.

    J Pharmacol Exp Ther 1999;289:800-6.

    [59] Serkova N, Litt L, Leibfritz D, et al. The novel immunosuppressant

    SDZ-RAD protects rat brain slices from cyclosporine-induced

    reduction of high-energy phosphates. Br J Pharmacol 2000;129:

    485 -92.

    [60] Kasiske B, Cosio FG, Beto J, et al. Clinical practice guidelines for

    managing dyslipidemias in kidney transplant patients: a report from

    the Managing Dyslipidemias in Chronic Kidney Disease Work

    Group of the National Kidney Foundation Kidney Disease Outcomes

    Quality Initiative. Am J Transplant 2004;4(Suppl 7):13 -53.

    [61] Mathis AS, Dave N, Knipp GT, et al. Drug-related dyslipidemia after

    renal transplantation. Am J Health Syst Pharm 2004;61:565-85.

    [62] Blum CB. Effects of sirolimus on lipids in renal allograft recipients:

    an analysis using the Framingham risk model. Am J Transplant

    2002;2:551 -9.

    [63] Teruel JL, Escobar EM, Quereda C, et al. A simple and safe method

    for management of lymphocele after renal transplantation. J Urol

    1983;130:1058-9.

    [64] Bischof G, Rockenschaub S, Berlakovich G, et al. Management

    of lymphoceles after kidney transplantation. Transpl Int 1998;11:

    277 -80.

    [65] Fuller TF, Kang SM, Hirose R, et al. Management of lymphoceles

    after renal transplantation: laparoscopic versus open drainage. J Urol

    2003;169:2022-5.

    [66] Flechner SM, Zhou L, Derweesh I, et al. The impact of sirolimus,

    mycophenolate mofetil, cyclosporine, azathioprine, and steroids on

    wound healing in 513 kidney-transplant recipients. Transplantation

    2003;76:1729-34.

    [67] Knight RJ, Villa M, Welsh M, et al. Risk factors for impaired wound

    healing in sirolimus treated renal transplant recipients. Am J

    Transplant 2003;3(Suppl 5):481.

    [68] Hariharan S, Peddi VR, Savin VJ, et al. Recurrent and de novo renal

    diseases after renal transplantation: a report from the renal allograft

    disease registry. Am J Kidney Dis 1998;31:928-31.

    [69] Chapman JR. Optimizing the long-term outcome of renal transplants:

    opportunities created by sirolimus. Transplant Proc 2003;35:67 -72.

    [70] Wilmer WA, Rovin BH, Hebert CJ, Rao SV, Kumor K, Hebert LA.

    Management of glomerular proteinuria: a commentary. J Am Soc

    Nephrol 2003;14:3217-32.

    [71] Muirhead N, House A, Hollomby DJ, et al. Effect of valsartan on

    urinary protein excretion and renal function in patients with chronic

    renal allograft nephropathy. Transplant Proc 2003;35:2412-4.

    [72] Mourad G, Vela C, Ribstein J, et al. Long-term improvement in renal

    function after cyclosporine reduction in renal transplant recipients

    with histologically proven chronic cyclosporine nephropathy. Trans-

    plantation 1998;65:661 -7.

    [73] Hausberg M, Barenbrock M, Hohage H, et al. ACE inhibitor versus

    b-blocker for the treatment of hypertension in renal allograftrecipients. Hypertension 1999;33:862-8.

    [74] Suwelack B, Kobelt V, Erfmann M, et al. Long-term follow-up of

    ACE-inhibitor versus beta-blocker treatment and their effects on

    blood pressure and kidney function in renal transplant recipients.

    Transpl Int 2003;16:313 -20.

  • J. Pascual et al. / Transplantation Reviews 20 (2006) 118 17[75] Lorenz M, Kletzmayr J, Perschl A, et al. Anemia and iron

    deficiencies among long-term renal transplant recipients. J Am Soc

    Nephrol 2002;13:794-7.

    [76] Winkelmayer WC, Kewalramani R, Rutstein M, et al. Pharmacoe-

    pidemiology of anemia in kidney transplant recipients. J Am Soc

    Nephrol 2004;15:1347-52.

    [77] Campistol JM, Gutierrez-Dalmau A, Torregrosa JV. Conversion to

    sirolimus: a successful treatment for posttransplantation Kaposis

    sarcoma. Transplantation 2004;77:760 -2.

    [78] Stallone G, Schena A, Infante B, et al. Sirolimus for Kaposis

    sarcoma in renal-transplant recipients. N Engl J Med 2005;352:

    1317-23.

    [79] Mathew T, Kreis H, Friend P. Two-year incidence of malignancy in

    sirolimus-treated renal transplant recipients: results from five

    multicenter studies. Clin Transplant 2004;18:446-9.

    [80] Pascual J, Marcen R, Ortuno J. Clinical experience with everolimus

    (Certican) in elderly recipients: the bold-for-oldQ concept. Transplan-tation 2005;79:S85-8.

    [81] Kahan BD. Efficacy of sirolimus compared with azathioprine for

    reduction of acute renal allograft rejection: a randomised multicentre

    study. The Rapamune US Study Group. Lancet 2000;356:194-202.

    [82] MacDonald AS. A worldwide, phase III, randomized, controlled,

    safety and efficacy study of a sirolimus/cyclosporine regimen for

    prevention of acute rejection in recipients of primary mismatched

    renal allografts. Transplantation 2001;71:271 -80.

    [83] Hoogeveen RC, Ballantyne CM, Pownall HJ, et al. Effect of

    sirolimus on the metabolism of apoB100-containing lipoproteins in

    renal transplant patients. Transplantation 2001;72:1244-50.

    [84] Tur MD, Garrigue V, Vela C, et al. Apolipoprotein CIII is upregulated

    by anticalcineurins and rapamycin: implications in transplantation-

    induced dyslipidemia. Transplant Proc 2000;32:2783-4.

    [85] Morrisett JD, Abdel-Fattah G, Hoogeveen R, et al. Effects of sirolimus

    on plasma lipids, lipoprotein levels, and fatty acid metabolism in renal

    transplant patients. J Lipid Res 2002;43:1170-80.

    [86] Eisen HJ, Tuzcu EM, Dorent R, et al. Everolimus for the prevention

    of allograft rejection and vasculopathy in cardiac-transplant recipi-

    ents. N Engl J Med 2003;349:847 -58.

    [87] Kovarik JM, Hartmann S, Hubert M, et al. Pharmacokinetic and

    pharmacodynamic assessments of HMG-CoA reductase inhibitors

    when coadministered with everolimus. J Clin Pharmacol 2002;

    42:222-8.

    [88] Jardine AG, Holdaas H, Fellstrom B, et al. Fluvastatin prevents

    cardiac death and myocardial infarction in renal transplant recipients:

    post-hoc subgroup analyses of the ALERT Study. Am J Transplant

    2004;4:988 -95.

    [89] Kahan BD, Podbielski J, Napoli KL, et al. Immunosuppressive

    effects and safety of a sirolimus/cyclosporine combination regimen

    for renal transplantation. Transplantation 1998;66:1040-6.

    [90] Lezaic V, Djukanovic L, Pavlovic-Kentera V. Recombinant human

    erythropoietin treatment of anemia in renal transplant patients. Ren

    Fail 1995;17:705-14.

    [91] Pascual J, Marcen R, Ortuno J. Clinical experience with everolimus

    (Certican): optimizing dose and tolerability. Transplantation 2005;

    79:S80-4.

    [92] Yatscoff RW, Fryer J, Thliveris JA. Comparison of the effect of

    rapamycin and FK506 on release of prostacyclin and endothelin in

    vitro. Clin Biochem 1993;26:409 -14.

    [93] Cahill BC, Somerville KT, Crompton JA, et al. Early experience with

    sirolimus in lung transplant recipients with chronic allograft

    rejection. J Heart Lung Transplant 2003;22:169 -76.

    [94] Kaplan MJ, Ellis CN, Bata-Csorgo Z, et al. Systemic toxicity

    following administration of sirolimus (formerly rapamycin) for

    psoriasis: association of capillary leak syndrome with apoptosis of

    lesional lymphocytes. Arch Dermatol 1999;135:553-7.

    [95] Citterlo F, Scata MC, Violi P, et al. Rapid conversion to sirolimus for

    chronic progressive deterioration of the renal function in kidney

    allograft recipients. Transplant Proc 2003;35:1292-4.[96] Mohaupt MG, Vogt B, Frey FJ. Sirolimus-associated eyelid edema in

    kidney transplant recipients. Transplantation 2001;72:162 -4.

    [97] Khauli RB, Stoff JS, Lovewell T, et al. Post-transplant lymphoceles:

    a critical look into the risk factors, pathophysiology and manage-

    ment. J Urol 1993;150:22-6.

    [98] Ciancio G, Burke GW, Gaynor JJ, et al. A randomized long-term trial

    of tacrolimus/sirolimus versus tacrolimus/mycophenolate mofetil

    versus cyclosporine (Neoral)/sirolimus in renal transplantation. II.

    Survival, function, and protocol compliance at 1 year. Transplanta-

    tion 2004;77:252-8.

    [99] Kandaswamy R, Melancon JK, Dunn T, et al. A prospective rando-

    mized trial of steroid-free maintenance regimens in kidney transplant

    recipientsan interim analysis. Am J Transplant 2005;5:1529 -36.

    [100] Napoli KL, Wang ME, Stepkowski SM, et al. Relative tissue

    distributions of cyclosporine and sirolimus after concomitant peroral

    administration to the rat: evidence for pharmacokinetic interactions.

    Ther Drug Monit 1998;20:123-33.

    [101] Podder H, Stepkowski SM, Napoli KL, et al. Pharmacokinetic

    interactions augment toxicities of sirolimus/cyclosporine combina-

    tions. J Am Soc Nephrol 2001;12:1059-71.

    [102] Andoh TF, Lindsley J, Franceschini N, et al. Synergistic effects of

    cyclosporine and rapamycin in a chronic nephrotoxicity model.

    Transplantation 1996;62:311 -6.

    [103] Bonegio R, Lieberthal W. Role of apoptosis in the pathogenesis of

    acute renal failure. Curr Opin Nephrol Hypertens 2002;11:301-8.

    [104] Keane WF. Proteinuria: its clinical importance and role in

    progressive renal disease. Am J Kidney Dis 2000;35:S97-S105.

    [105] Morelon E, Kreis H. Sirolimus therapy without calcineurin inhibitors:

    Necker Hospital 8-year experience. Transplant Proc 2003;35:52 -7.

    [106] Coombes J, Mreich E, Liddle C, Rangan GK. Rapamycin worsens

    renal function and intratubular cast formation in protein-overload

    nephropathy. Kidney Int 2005;68:2599-607.

    [107] Vanrenterghem Y, Ponticelli C, Morales JM, et al. Prevalence and

    management of anemia in renal transplant recipients: a European

    survey. Am J Transplant 2003;3:835 -45.

    [108] Cohen B, Smits JM, Haase B, et al. Expanding the donor pool to

    increase renal transplantation. Nephrol Dial Transplant 2005;20:34-41.

    [109] Eurotransplant. Eurotransplant Annual Reports 19962003.

    www.eurotransplant.nl/?id= annual_report [Last accessed: June

    2005].

    [110] Morrissey PE, Gohh R, Yango A, et al. Renal tra