Everolimus and Long-Term Outcomes in Renal Transplantation
Josep M. Campistol,Johan W. de Fijter,Björn Nashan,Hallvard Holdaas,Štefan Vı́tko,Christophe Legendre
DOI: https://doi.org/10.1097/tp.0b013e3182230900
2011-01-01
Transplantation
Abstract:SECTION 1: LONG-TERM OUTCOMES IN RENAL TRANSPLANTATION AND THE RATIONALE FOR EARLY USE OF EVEROLIMUS Josep M. Campistol A series of recent meetings brought together experts in the field of transplantation to discuss the challenge of improving long-term outcomes and establishing optimal strategies for immunosuppression after renal transplant. This supplement is a summary of these meetings, the objectives of which were to present the supporting data and rationale for using the mammalian target of rapamycin inhibitor (mTORi) everolimus as a part of a preemptive strategy to improve long-term outcomes, and to discuss practical aspects and proposed algorithms for preemptive use of everolimus. Long-Term Outcomes in Renal Transplantation Over the past 20 years, the rate of acute rejection after renal transplant has improved significantly as a result of the introduction of new immunosuppressant drugs such as cyclosporine A (CsA), mycophenolate mofetil (MMF), and tacrolimus (1). However, there has been little or no improvement in long-term outcomes for the recipients of renal transplants (2, 3) for two main reasons: chronic allograft nephropathy (CAN)/interstitial fibrosis and tubular atrophy (IFTA) and death with a functioning graft. CAN/IFTA CAN/IFTA is a pathophysiologic entity that accounts for one third of renal transplant graft failures (4). Changes in renal pathology associated with CAN/IFTA include IFTA, arteriolar hyalinosis, arteriosclerosis, and glomerulosclerosis, which are accompanied by indicators of a progressive decline in graft function such as increasing plasma creatinine, proteinuria, and hypertension (5, 6). There are multiple factors that can contribute to the development of CAN/IFTA, including an acute rejection episode within 1 year of transplant, delayed graft function (DGF), and infection, although the most significant factor is the use of calcineurin inhibitor (CNI) therapy. CNI-associated nephrotoxicity occurs early after transplant and progresses over time, including striped cortical fibrosis and tubular microcalcifications in addition to the development of arteriolar hyalinosis. Most patients exhibit signs of nephrotoxicity within 1 year of transplant (7, 8). Data have demonstrated that lowering CNI dose can reduce CNI nephrotoxicity (8); therefore, it has been proposed that the duration of CNI therapy should be limited, if possible (9, 10). Another factor that contributes to graft loss is viral infection, particularly cytomegalovirus (CMV) and BK virus (BKV). CMV infection increases the risk of acute rejection and is implicated in the development of CAN/IFTA (11) because of inflammation and stimulation of the immune response; this causes further graft damage and eventually graft loss (12, 13). BKV reactivation causes BKV nephropathy, which leads to graft loss in 50% of cases 6 to 60 months posttransplant (14). Death With a Functioning Graft Approximately 50% of graft loss in transplant recipients occurs in patients with a functioning graft (4). The main causes of death in these cases are cardiovascular disease (CVD) and malignancy. Patients with chronic renal dysfunction and end-stage renal disease are often at risk of developing CVD before transplant, and this is exacerbated by immunosuppressant drugs after transplant (15). CNIs, in particular, can contribute to the development of CVD by increasing arteriolar vasoconstriction. Furthermore, CNI therapy can increase the risk of developing diabetes and atherosclerosis, which are also the risk factors for CVD events (15). Malignancy is the second highest cause of death in transplant patients with a functioning graft; there is a 3-fold higher risk of developing cancer compared with the normal population (16). CNI therapy has been associated with increased risk of malignancy: immunosuppressive agents impair the normal immunosurveillance mechanisms that would detect malignant cells (16), and CsA has been shown to induce double-strand DNA breaks and inhibit DNA repair mechanisms (17, 18). As CNI therapy is known to increase the risk of CAN/IFTA, CVD, and malignancy, which all contribute to poor long-term outcomes, there is a rationale for reducing the exposure to CNIs (19). Options for CNI Tapering The mTORis everolimus and sirolimus offer the potential for reducing CNI exposure. These immunosuppressant drugs block mTOR, preventing downstream signaling including DNA replication and T-cell proliferation. The CONVERT study investigated the potential of late CNI elimination (6 to 120 months after transplant) with conversion to an mTORi maintenance therapy to improve renal function in kidney transplant patients. Data demonstrated that patients who were already showing signs of renal damage at the beginning of the study experienced no improvement in renal function, whether they were switched to sirolimus or not (20). The study concluded that once signs of nephrotoxicity are apparent, reducing or eliminating CNI therapy does not improve outcomes. Therefore, to minimize CNI-induced nephrotoxicity, it is vital to reduce or stop CNI therapy before the signs of renal damage appear. Several studies have demonstrated the potential for using preemptive everolimus to eliminate or minimize CNI therapy, with good efficacy and comparable or improved renal function (21–25). Clinical trials investigating everolimus with reduced CsA have shown good efficacy and comparable or improved renal function with CsA reduction (23, 25, 26). In addition, the antiproliferative effects of mTORis may provide other potential benefits: a low incidence of adverse events commonly associated with CNIs; a reduced incidence of infections, including CMV and BKV; and fewer de novo malignancies (19, 20, 24, 25, 27). This supplement has been prepared to summarize the rationale and the data to provide guidance for the use of preemptive everolimus in renal transplant patients. Section 2 describes the mode of action and clinical profile of everolimus in detail. The next two sections then discuss the supporting data for, and provide practical guidance on, preemptive CNI minimization with everolimus and early conversion to everolimus with CNI elimination (Fig. 1). The practical guidance will also be illustrated with profiles of patients who could be considered suitable for these strategies. Section 5 describes the ongoing clinical trials with preemptive everolimus, which seek to address some of the unmet needs of renal transplant patients.FIGURE 1.: Schematic of the preemptive therapy strategies. CNI, calcineurin inhibitor.SECTION 2: EVEROLIMUS: MODE OF ACTION AND CLINICAL PROFILE Johan W. de Fijter, Štefan Vítko Introduction to Everolimus Everolimus is an mTORi and a potent orally active immunosuppressant that is derived from a macrocyclic lactone (28). mTORis have a mode of action that is distinct from those of other immunosuppressants, such as CNIs and inosine monophosphate dehydrogenase pathway inhibitors (29, 30). CNIs suppress early T-cell activation by inhibiting the transcription of T-cell-specific genes, such as interleukin (IL)-2. Inosine monophosphate dehydrogenase inhibitors inhibit T-cell proliferation more generally by inhibiting DNA synthesis by blocking the lymphocyte-specific de novo purine synthesis pathway. Everolimus and sirolimus, instead, inhibit the IL-2-stimulated T-cell proliferation pathway, in which mTOR is a central component. mTOR is a serine/threonine kinase that regulates cell growth, cell cycle progression, and proliferation signaling pathways that respond to environmental and nutritional conditions and hence has a pleiotropic role in physiologic processes (31, 32). mTORis block growth factor-driven proliferation of both hematopoietic and nonhematopoietic cells, including vascular and bronchial smooth muscle cells, and their antiproliferative effects are not limited to the immune system (Fig. 2) (33, 34). By binding mTOR, everolimus inhibits the activation of p70S6 kinase, which arrests the cell cycle at the G1 to S phase. Nanomolar concentrations of the drug can inhibit IL-2 signaling pathways.FIGURE 2.: Mammalian target of rapamycin (mTOR) pathway and inhibition: pleiotropic effects. Data obtained from Ponticelli, Hudes, Guba et al., Buell et al., Contreras et al., Cruzado et al., Eisen, Ormiston et al., and Schuler et al. (29, 53, 62, 94–99). CMV, cytomegalovirus; CAN, chronic allograft nephropathy; CAV, cardiac allograft vasculopathy.Pharmacokinetics The chemical structure of everolimus differs from that of another mTORi, sirolimus, by the addition of 2-hydroxyethyl group at C40 (35, 36). This results in differences in the pharmacokinetic and pharmacodynamic properties of the two drugs (37, 38). Everolimus has a shorter half-life than sirolimus (28 vs. 62 hr, respectively): no loading dose is required and it reaches steady state more quickly than sirolimus. Both everolimus and CsA are metabolized by the cytochrome P450 isozyme CYP3A and are substrates for P-glycoprotein and, therefore, have the potential for interaction. It has been demonstrated that CsA increases levels of everolimus, but no such interaction has been seen with tacrolimus. In practice, this means that higher doses of everolimus are required to reach target levels when it is used in combination with tacrolimus than with CsA (39, 40). Co-administration of everolimus with strong CYP3A4 inhibitors and inducers, such as ketoconazole, itraconazole, ritonavir, and rifampicin, is not recommended. Efficacy: Acute Rejection and Renal Function Early studies demonstrated that combining everolimus (1.5 or 3.0 mg/day) with standard-exposure CsA provided equivalent efficacy against acute rejection as the standard combination of CsA and MMF; however, renal function was inferior (25, 41). Subsequent studies have investigated the role of everolimus with CNI minimization or elimination to reduce CNI toxicities, particularly nephrotoxicity, while maintaining efficacy against acute rejection. Several studies have investigated everolimus-based regimens combined with CNI minimization (Table 1). Two prospective, randomized studies of everolimus 1.5 or 3.0 mg/day (everolimus trough level >3 ng/mL) with low CsA in de novo renal transplant recipients were undertaken, using C2 monitoring of CsA exposure (the 2-hr postdose blood CsA concentration) and demonstrated that this was a feasible strategy (42). The incidence of efficacy failure was comparable with that of standard CsA regimens; this was achieved with approximately 60% reduction at 12 months in CsA level.TABLE 1: Everolimus facilitates CsA minimization without compromising efficacyTo test these conclusions, the efficacy and safety of everolimus in combination with low CsA have been compared directly with the standard CsA immunosuppressive regimen in the A2309 study (24). As described more fully elsewhere in this supplement, the study demonstrated more than 60% reduction in CsA levels at 12 months compared with mycophenolic acid (MPA) and standard CsA, with no compromise in biopsy-proven acute rejection (BPAR) rates (Fig. 3) or safety.FIGURE 3.: Everolimus with calcineurin inhibitor (CNI) minimization provides similar efficacy to standard CNI strategies. BPAR, biopsy-proven acute rejection; CsA, cyclosporine A; MPA, mycophenolic acid.Another option is to eliminate CsA from the immunosuppressive regimen using everolimus at an early time point posttransplant. Several studies of everolimus with early CNI elimination, which show significant improvement in renal function versus CNI continuation, will be discussed in more detail elsewhere in this supplement (see Section 4). These data demonstrate that CNI minimization or elimination in combination with an mTORi, such as everolimus, has the potential to reduce CNI nephrotoxicity without increasing the risk of acute rejection (22, 25, 26, 42, 43) and could be of particular significance to patient populations that are at a greater risk of CNI-related side effects, such as those with CAN/IFTA or declining graft function, of an advanced age (>50 years), and of African-American ethnicity. Other Potential Benefits of Everolimus Reduction of CAN/IFTA and Chronic Allograft Dysfunction Chronic allograft dysfunction and loss occur most frequently as a consequence of CAN/IFTA, which, in turn, is a complex, multifactorial process where the primary causes are acute rejection, vascular remodeling, CNI-induced nephrotoxicity, and CMV infection (11, 44, 45). Everolimus has the potential to ameliorate many of the factors that contribute to the development of CAN/IFTA, primarily by enabling CNI minimization or elimination and its additional effects on excess vascular remodeling, proliferation, and CMV infection (Fig. 2). Together these effects could inhibit the development of CAN/IFTA and improve long-term outcomes in renal transplantation (46). The antiproliferative effects of everolimus delayed the progression of CAN/IFTA when initiated at an early stage of development and ameliorated the condition when used at an advanced stage of CAN/IFTA in a preclinical model (47). In another preclinical study in sensitized nude rats, everolimus initiated 5 weeks posttransplant prolonged allograft survival with no signs of IFTA (48). These data support the hypothesis that preemptive use of everolimus before the development of CAN/IFTA, or at the early stages of CAN/IFTA, may play a role in preserving allograft function. Antiviral Properties CMV and BKV infections are common in transplant patients and can cause graft loss: CMV is a major factor in the development of CAN/IFTA, and BKV can cause nephropathy. Everolimus reduces the incidence of both CMV and BKV (24, 25, 49). At 12 months in Study B201, the incidence of CMV infection in patients treated with everolimus plus CsA was significantly lower at 5.0% compared with 19.4% in the MMF plus CsA group (P=0.001; Fig. 4a). This was maintained at 36 months (5.7% vs. 19.9%, respectively) (25, 49). Similarly, in Study A2309, the incidence of CMV at 12 months was much lower in the everolimus-treated patients than in those treated with MMF (0.7%, 0.0%, and 5.9% for 1.5 mg everolimus, 3.0 mg everolimus, and MMF; Fig. 4b) (24). The incidence of BKV at 12 months in Study A2309 was reduced by everolimus plus low CsA compared with MPA plus standard CsA (0.7% to 1.1% vs. 1.8%, respectively; Fig. 4b).FIGURE 4.: (a) Everolimus significantly reduced the incidence of cytomegalovirus (CMV) infection in Study B201; (b) everolimus significantly reduced the incidence of CMV and BK virus (BKV) infection in Study A2309. CsA, cyclosporine A; MMF, mycophenolate mofetil; MPA, mycophenolic acid.CVD The renal transplant population is at a higher risk of a CVD event than the general population (15, 50, 51), and CVD is a major cause of death with a functioning graft (4). Although there are limited data regarding the cardiovascular benefits of everolimus in renal transplantation (52), evidence from heart transplant patients is promising. The antiproliferative effects of everolimus in combination with its antiangiogenic and vascular-remodeling properties have cardiovascular benefits, reducing the incidence of cardiac allograft vasculopathy, coronary artery intimal proliferation, and major adverse cardiac events compared with azathioprine in cardiac transplant recipients (Study B253) (53, 54). This trial demonstrated a lower incidence of cardiac allograft vasculopathy with everolimus compared with azathioprine, steroids, and standard CsA (35.7% vs. 52.8%, respectively; P=0.045). In addition, the change in maximal intimal thickness was 0.04 mm in the everolimus 1.5 mg group compared with 0.10 mm in the azathioprine group (P=0.01). Furthermore, the incidence of major adverse cardiac events after 4 years was 8.6% in the everolimus 1.5 mg group versus 15.3% in the azathioprine group. The antiproliferative effects of everolimus have also been exploited in the development of everolimus-eluting stents for reducing in-stent neo-intimal hyperplasia and restenosis in coronary artery disease (55–61). The everolimus-eluting stents have been shown to be effective and well tolerated in three key studies (FUTURE I and II, and SPIRIT I). Antimalignancy Benefits In addition to CVD, malignancies are one of the most common causes of death with a functioning graft, and transplanted patients are at an increased risk of developing cancer (62). The use of immunosuppressive agents may have a pathogenic role in the development of malignancies: immunosuppressive agents impair the body's immunosurveillance mechanisms that would normally monitor and prevent the growth of malignancies (16). More specifically, preclinical data have demonstrated that CsA inhibits normal DNA repair mechanisms and can induce double-stranded DNA breaks, thus allowing mutations to accumulate (17, 18, 63, 64). Indeed, transplanted patients are associated with an increased risk of developing skin cancer (melanoma, 2.2-fold higher; non-melanoma, 2.6-fold higher), non-Hodgkin's lymphoma (3.3-fold higher), Kaposi sarcoma (9.0-fold higher), and cancer of the kidney (1.4-fold higher) when compared with waiting-list patients (16, 65). mTORis have antimalignancy effects as a result of their inhibition of cellular signaling pathways involved in cell division and growth factor production. There is a solid body of evidence from both preclinical and clinical studies on the antimalignancy activity of everolimus (66–69). In preclinical studies, mTOR inhibition has been shown to induce apoptosis of epithelial cells and reverse Akt-dependent neoplasia (69), reducing tumor volume, inhibiting tumor growth, and sensitizing tumor cells to DNA damage-induced apoptosis (66, 67). In a retrospective analysis using registry data of renal transplant patients, there was a reduced incidence of de novo malignancies with mTORi-containing regimens compared with those that contained CNIs (Table 2) (20, 27). A2309, the largest randomized study of everolimus, has also demonstrated reduced malignancies for everolimus: 3.3% of patients developed a neoplasm compared with 5.9% in the MPA group (24). Independent of its indication in transplantation, everolimus has been developed as a treatment for cancer and has been approved for second-line therapy of metastatic renal cell carcinoma in both the United States and all 27 countries of the European Union. In this indication, the RECORD study demonstrated doubling of progression-free survival compared with placebo (4.0 vs. 1.9 months) in patients whose metastatic disease had progressed on other targeted therapies (70).TABLE 2: Reduced incidence of de novo malignancies with mTORi-containing regimens compared with CNI-containing regimens (27)Common mTORi-Related Adverse Events and Their Management The transplant population usually has high rates of adverse events as a result of surgery and treatment regimens. mTORis as a class, however, are associated with a number of specific adverse events, such as proteinuria, dyslipidemia, wound-healing events, and edema, which are considered to be dose dependent (71–73). In general, proteinuria seems to be associated with conversion of maintenance patients to mTORis and those with existing proteinuria, rather than with de novo mTORi use (74–76). As such, conversion to everolimus is not advised in patients with preexisting proteinuria more than 800 mg/day (77). It has been suggested that proteinuria associated with mTORi can be managed by the initiation of an angiotensin-converting enzyme inhibitor and/or angiotensin receptor blocker (78–80). The incidence of edema observed with everolimus is comparable with that seen with MPA (81). Edema can be treated using loop diuretics (e.g., furosemide), and everolimus levels can be maintained in mild cases, although lowering everolimus doses should be considered (82). Recent data suggest the incidence of hyperlipidemia associated with everolimus is also similar to that seen with MPA (81). In cases of dyslipidemia, these can be treated simply with statins (49, 83–85). However, if the response to statins is insufficient, reduction in everolimus levels or conversion from CsA to tacrolimus may also need to be considered (81, 86). Other adverse events include anemia and arthralgia, which are dose independent. If hyperlipidemia, lymphocele, or wound-healing complications develop, these events generally occur at initiation of mTORi treatment (87). The development of CAN/IFTA, proteinuria, hypertension, anemia, and malignancy are usually associated with maintenance therapy. Most adverse events associated with everolimus therapy are generally mild and can be managed by basic intervention (Table 3). The safety profile of everolimus with low CsA compares favorably with full-dose CNI regimens (42).TABLE 3: Common side effects of everolimus and management strategiesThere have been some concerns that, as has been observed with sirolimus, the early introduction of everolimus may increase the incidence of DGF and wound-healing events (88–92). However, the results of a randomized, multicenter trial demonstrated that kidney function recovery, wound healing (approximately 40%), efficacy, and tolerance at 3 months posttransplant were similar when everolimus was administered immediately after kidney transplant or when therapy was delayed for 4 weeks (93). Taken together with wound-healing data from Study A2309 (24), which showed similar wound-healing events in the everolimus and MMF arms (35% and 39% for 1.5 and 3.0 mg everolimus, respectively, and 26% for MMF), the concerns over wound healing with everolimus should not preclude its use. Conclusions mTOR regulates cell growth and proliferation signaling pathways that respond to environmental and nutritional conditions and has a role in many different physiologic processes, including T-cell proliferation. By inhibiting mTOR, everolimus has pleiotropic effects that result in effective immunosuppression and can help to improve long-term outcomes. Indeed, there is growing clinical evidence that it can inhibit development of CAN/IFTA, CMV and BKV infection, and malignancy. Everolimus has a complementary mechanism of action to CNIs. This enables it to be combined with reduced-exposure CNIs, thus decreasing the risk of nephrotoxicity and other CNI adverse events without compromising BPAR rates. There is a growing body of evidence that preemptive introduction of everolimus (before month 6) with CNI minimization or elimination improves renal function at 1 year, an early indicator of long-term allograft function. Adverse events associated with everolimus tend to be mild and can be managed with basic interventions. SECTION 3: EXPERIENCE AND CONSIDERATIONS FOR CNI MINIMIZATION WITH EVEROLIMUS Björn Nashan, Christophe Legendre, Štefan Vítko Although CNIs, CsA, and tacrolimus improve acute rejection rates and allograft survival in the first year posttransplant, they are associated with nephrotoxicity and development of CAN/IFTA, a leading cause of graft loss in kidney transplant patients (8, 45). As such, recent focus has been placed on minimizing exposure to CNIs to reduce nephrotoxicity and improve long-term outcomes, in particular, the timing of minimization to optimize the benefits of reducing CNI exposure while controling rejection in the critical period posttransplant. Because CNI nephrotoxicity occurs soon after therapy, with the majority of patients demonstrating signs of nephrotoxicity within 1 year of transplant (8), there is an obvious advantage for reducing CNI exposure early posttransplant. There are a number of approaches to reduce CNI exposure: CNI avoidance, CNI elimination, and CNI minimization. Rationale for CNI Minimization In a single-arm, CNI-avoidance study, in which patients were given de novo daclizumab, MMF, and corticosteroid therapy, a 48% BPAR rate was reported (100). Daclizumab is a humanized monoclonal antibody that binds to IL-2 receptors and, thus, inhibits T-cell proliferation. Although adequate blockade of the IL-2 receptor was achieved in this study, it was noted that alternative T-cell activation pathways, such as the IL-15 receptor-mediated pathway, may also have played a role in acute rejection (101, 102). This suggests that CNIs are still required to avoid acute rejection, even in low-risk patients, such that a CNI minimization approach would be a more appropriate option. CNI minimization as a potential strategy for reducing CNI exposure was investigated in a recent large three-arm study of de novo CNI minimization versus de novo CNI minimization and subsequent CNI elimination or standard CsA. The 12-month CAESAR study showed that mean glomerular filtration rate (GFR) and incidence of BPAR rates were similar with CsA minimization and standard CsA (50.9 vs. 48.6 mL/min/1.73 m2 for GFR; 27.5% vs. 25.4% for BPAR, respectively) (103), demonstrating that de novo CNI minimization is a feasible option. Against this background, everolimus, an mTORi, has demonstrated synergistic immunosuppressive effects with CNIs in preclinical models (104). Furthermore, as the IL-15 signaling pathway, which is also associated with ongoing acute rejection (101, 102) and T-cell proliferation, is mediated by mTOR (105), there is a strong rationale for including everolimus in a CNI minimization regimen. Based on these assumptions, the CNI minimization trial B156 was designed and initiated in 1998 (23); data from this study established the potent synergism of an mTORi and low CNI regimen, which has also been demonstrated in subsequent trials (discussed later in this section). These data suggest that everolimus may be of use in CNI minimization protocols for improving long-term outcomes while maintaining efficacy against acute rejection. The role of everolimus in CNI elimination strategies will be discussed elsewhere in this supplement (see Section 4). Early Studies: Development of Everolimus in the De Novo Setting A number of key clinical studies have demonstrated the efficacy of everolimus in renal transplantation in the de novo setting. The first was B156, a 36-month, randomized, open-label, phase II study of everolimus 3.0 mg/day in combination with standard or low CsA (23). At 12 months, efficacy failure (composite endpoint of BPAR, graft loss, death, and loss to follow-up) was significantly lower for the low- versus standard-CsA group (8.6% vs. 28.3%; P=0.012), with improved renal function reported in the low-CsA group (creatinine clearance: 61 vs. 54 mL/min; P=0.007). Treatment with low CsA was also associated with a better safety profile, suggesting that de novo everolimus with low CsA provides effective immunosuppression while minimizing nephrotoxicity (23). At a similar time, two pivotal registration studies—B201 and B251—were investigating a different de novo strategy, whereby everolimus (1.5 or 3.0 mg/day) was administered with standard CsA and corticosteroids, and compared with the standard immunosuppressive regimen (25, 41). As with Study B156, de novo everolimus was associated with comparable efficacy to MMF in terms of incidence of efficacy failure and BPAR but, with standard CsA, this was at the expense of reduced renal function at 1 year. Protocol amendments, based on B156 and the initial findings from B201 and B251, to reduce CsA C0 levels were introduced and renal function improved (23, 25, 41). Interestingly, in Study B201, although the proportions of patients receiving CMV prophylaxis were similar, more patients in the MMF group had CMV infection than either of the everolimus groups (19.4% vs. 5.2% and 7.6% for MMF, everolimus 1.5 [P=0.0006], and 3.0 mg/day [P>0.0001], respectively) (49). Furthermore, an analysis of patients at high risk of CMV reactivation (CMV-positive donor/CMV-negative recipient) demonstrated that the incidence of CMV reactivation was approximately doubled for patients receiving MMF than for patients receiving everolimus (1.5 or 3.0 mg), regardless of whether antiviral prophylaxis was administered (Fig. 5). These data suggest that everolimus may also have antiviral properties, which may prevent CAN/IFTA associated with CMV infection, and having antimitotic, antimalignancy, and immunosuppressive functions.FIGURE 5.: B201: incidence of cytomegalovirus (CMV) infections in renal transplant patients at high risk of CMV reactivation receiving everolimus or mycophenolate mofetil (MMF) with or without antiviral prophylaxis.Based on the data from B156, B201, and B251, two 12-month, randomized, phase III studies (A2306 and A2307) were designed and conducted to compare the efficacy and safety of de novo everolimus (1.5 or 3.0 mg/day) in combination with low CsA (in this case, guided by concentration 2 hr after administration [C2] monitoring). An analysis of the CsA levels from these trials at 12 months confirmed that everolimus can facilitate early CNI minimization (Fig. 6). Efficacy results showed that the composite efficacy endpoint (defined as occurrence of BPAR, graft loss, death, or loss to follow-up) was similar for both everolimus groups, although induction therapy was associated with reduced incidence of overall rejection (26). Renal function was also preserved and was similar between treatment groups, thus confirming that de novo everolimus with CNI minimization is a valid immunosuppressive option.FIGURE 6.: Everolimus can facilitate significant calcineurin inhibitor minimization (Novartis data on file). CsA, cyclosporine A; MMF, mycophenolate mofetil.De Novo Everolimus With CsA Minimization: Study A2309 Study A2309 is the largest single-registration study using everolimus ever undertaken in renal transplantation and has evaluated the efficacy and safety of de novo everolimus with low CsA as a strategy for improving long-term renal graft outcomes (24).