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).
What problem does this paper attempt to address?
-
Everolimus and Sirolimus in Transplantation-Related but Different.
Jost Klawitter,Björn Nashan,Uwe Christians
DOI: https://doi.org/10.1517/14740338.2015.1040388
2015-01-01
Expert Opinion on Drug Safety
Abstract:INTRODUCTION:The inhibitors of the mammalian target of rapamycin (mTOR) sirolimus and everolimus are used not only as immunosuppressants after organ transplantation in combination with calcineurin inhibitors (CNIs) but also as proliferation signal inhibitors coated on drug-eluting stents and in cancer therapy. Notwithstanding their related chemical structures, both have distinct pharmacokinetic, pharmacodynamic and toxicodynamic properties.AREAS COVERED:The additional hydroxyethyl group at the C(40) of the everolimus molecule results in different tissue and subcellular distribution, different affinities to active drug transporters and drug-metabolizing enzymes as well as differences in drug-target protein interactions including a much higher potency in terms of interacting with the mTOR complex 2 than sirolimus. Said mechanistic differences as well as differences found in clinical trials in transplant patients are reviewed.EXPERT OPINION:In comparison to sirolimus, everolimus has higher bioavailability, a shorter terminal half-life, different blood metabolite patterns, the potential to antagonize the negative effects of CNIs on neuronal and kidney cell metabolism (which sirolimus enhances), the ability to stimulate mitochondrial oxidation (which sirolimus inhibits) and to reduce vascular inflammation to a greater extent. A head-to-head, randomized trial comparing the safety and tolerability of these two mTOR inhibitors in solid organ transplant recipients is merited.
-
Efficacy And Safety Of Everolimus For Maintenance Immunosuppression Of Kidney Transplantation: A Meta-Analysis Of Randomized Controlled Trials
Jinyu Liu,Dong Liu,Juan Li,Lan Zhu,Chengliang Zhang,Kai Lei,Qiling Xu,Ruxu You
DOI: https://doi.org/10.1371/journal.pone.0170246
IF: 3.7
2017-01-01
PLoS ONE
Abstract:Background Conversion to everolimus is often used in kidney transplantation to overcome calcineurin inhibitor (CNI) nephrotoxicity but there is conflicting evidence for this approach.Objectives To investigate the benefits and harm from randomized clinical trials (RCTs) involving the conversion from CNI to everolimus after kidney transplantation.Methods Databases were searched up to March 2016. Two reviewers independently assessed trials for eligibility and quality, and extracted data.Results are expressed as risk ratio (RR) or mean difference (MD) with 95% confidence intervals (CI). Results Eleven RCTs, with a total of 1,633 patients, met the final inclusion criteria. Patients converted to everolimus had improved renal function at 1 year posttransplant with an estimated glomerular filtration rate (eGFR) of 5.36 mL/min per 1.73 m(2) greater than patients remaining on CNI (p = 0.0005) and the longer-term results (> 1 year) of renal function was identical to that of 1 year. There was not a substantial difference in graft loss, mortality, and the occurrence of adverse events (AEs) or serious AEs. However, the risks of acute rejection and trial termination due to AEs with everolimus are respectively 1.82 and 2.63 times greater than patients staying on CNI at 1 year posttransplant (p = 0.02, p = 0.03, respectively). Further, those patients who converted to everolimus had a substantially greater risk of anemia, hyperlipidemia, hypercholesterolemia, hypokalemia, proteinuria, stomatitis, mouth ulceration, and acne.Conclusions Conversion from CNI to everolimus after kidney transplantation is associated with improved renal function in the first 5 years posttransplant but increases the risk of acute rejection at 1 year posttransplant and may not be well endured.
-
Phase Ii Study Of Everolimus In Patients With Metastatic Colorectal Adenocarcinoma Previously Treated With Bevacizumab-, Fluoropyrimidine-, Oxaliplatin-, And Irinotecan-Based Regimens
Kimmie Ng,Josep Tabernero,Jimmy Hwang,Emilio Bajetta,Sunil Sharma,Salvatore A. Del Prete,Edward R. Arrowsmith,David P. Ryan,Michaela Sedova,Jin Jin,Kamel Malek,Charles S. Fuchs
DOI: https://doi.org/10.1158/1078-0432.CCR-13-0027
IF: 13.801
2013-01-01
Clinical Cancer Research
Abstract:Purpose: Dysregulation of the phosphoinositide 3-kinase (PI3K)/Akt/mTOR pathway is seen in 40% to 60% of patients with colorectal cancer. Everolimus, an oral inhibitor of mTOR, showed efficacy in patients with metastatic colorectal cancers in phase I studies.Experimental Design: In sequential phase II studies assessing two dosing schedules, patients with metastatic colorectal cancers refractory to bevacizumab-, fluoropyrimidine-, oxaliplatin-, and irinotecan-based regimens received everolimus 70 mg/wk (n = 99) or 10 mg/d (n = 100). Primary endpoints were disease control rate (DCR) and objective response rate; secondary endpoints included progression-free survival (PFS), overall survival (OS), and duration of response or stable disease (SD). Tumor tissue was collected from all patients for predefined exploratory biomarker analyses.Results: Seventy-one patients were included in the per-protocol set for each cohort. DCRs of 31.0% and 32.4% (all SD) were seen in the weekly and daily schedules, respectively. Median duration of SD was 3.9 months in each cohort. Median PFS and OS were 1.8 and 4.9 months and 1.8 and 5.9 months, respectively, for the weekly and daily schedules. Among patients receiving daily everolimus, those with a KRAS mutation experienced significantly shorter median OS (P = 0.008) and lower DCR (P = 0.035) compared with those with wild-type KRAS in exploratory biomarker analyses.Conclusions: Everolimus 70 mg/wk or 10 mg/d was well tolerated but did not confer meaningful efficacy in heavily pretreated patients with metastatic colorectal cancers. Future studies may consider evaluating everolimus in combination with other agents or in patients with dysregulation of the PI3K/Akt/mTOR pathway. (C) 2013 AACR.
-
Inhibitors of mTOR and risks of allograft failure and mortality in kidney transplantation.
T Isakova,H Xie,S Messinger,F Cortazar,J J Scialla,G Guerra,G Contreras,D Roth,G W Burke,M Z Molnar,I Mucsi,M Wolf
DOI: https://doi.org/10.1111/j.1600-6143.2012.04281.x
IF: 9.369
2013-01-01
American Journal of Transplantation
Abstract:Data on long-term outcomes of users of inhibitors of the mammalian target of rapamycin (mTORI) are lacking in kidney transplantation. In an analysis of 139 370 US kidney transplant recipients between 1999 through 2010, we compared clinical outcomes among users of mTORIs versus calcineurin inhibitors (CNI) in their primary immunosuppresive regimen. During the first 2 years posttransplantation, primary use of mTORIs without CNIs (N = 3237) was associated with greater risks of allograft failure and death compared with a CNI-based regimen (N = 125 623); the hazard ratio (HR) of the composite outcome ranged from 3.67 (95% confidence interval [CI], 3.124.32) after discharge to 1.40 (95% CI 1.261.57) by year 2. During years 28, primary use of mTORIs without CNIs was independently associated with greater risks of death (HR 1.25; 95% CI, 1.111.41) and the composite (HR 1.17; 95%CI, 1.081.27) in fully adjusted analyses. The results were qualitatively unchanged in subgroups defined by medical history, immunological risk and clinical course during the index transplant hospitalization. In a propensity-score matched cohort, use of mTORIs was associated with significantly worse outcomes during the first 2 years and greater risks of death (HR 1.21; 95% CI, 1.051.39) and the composite (HR 1.18; 95% CI, 1.081.30) in years 28. Compared with CNI-based regimens, use of an mTORI-based regimen for primary immunosuppression in kidney transplantation was associated with inferior recipient survival.
-
Everolimus in de novo liver transplant recipients: a systematic review.
Cheng-Yong Tang,Ai Shen,Xu-Fu Wei,Qing-Dong Li,Rui Liu,He-Jun Deng,Yong-Zhong Wu,Zhong-Jun Wu
DOI: https://doi.org/10.1016/S1499-3872(15)60419-2
IF: 3.355
2015-01-01
Hepatobiliary & Pancreatic Diseases International
Abstract:Everolimus has no nephrotoxicity and is used to treat patients with post-liver transplant chronic renal insufficiency. The present systematic review was to evaluate the efficacy and safety of everolimus in de novo liver transplant patients.Randomized controlled trials comparing everolimus for de novo liver transplant in PubMed, the Cochrane Library, and ScienceDirect published up to March 31, 2014 were searched by two independent reviewers. Mean differences and 95% confidence interval (95% CI) for renal function, relative risk (RR) and 95% CI for treated biopsy-proven acute rejection (tBPAR), graft loss, death, neoplasms/tumor recurrence, and adverse events were collected. Meta-analyses were performed with RevMan version 5.10.A total of four randomized controlled trials covering 1119 cases were included. The meta-analyses revealed that compared with standard exposure of calcineurin inhibitors (CNIs), everolimus combined with reduced CNIs improved creatinine clearance (calculated with the Cockcroft-Gault formula) by 5.13 mL/min at one year (95% CI: 0.42-9.84; P=0.03), and decreased tBPAR (RR: 0.56; 95% CI: 0.35-0.90; P=0.02). Everolimus initiation with CNIs elimination improved glomerular filtration rate (GFR, measured with the modification of diet in renal disease formula) of 10.42 mL/min/1.73 m2 (95% CI: 3.44-17.41; P<0.01) one year after treatment, but increased tBPAR (RR: 1.71; 95% CI: 1.15-2.53; P<0.01). Everolimus decreased the risk of neoplasms/tumor recurrence after liver transplant (RR: 0.60; 95% CI: 0.34-1.03; P=0.06), but was associated with greater risk of adverse events which resulted in drug discontinuation (RR: 1.98; 95% CI: 1.49-2.64; P<0.01).Early introduction of everolimus combined with low-dose or no CNI in de novo liver transplant significantly improves renal function one year post treatment. Everolimus combined with low-dose CNI decreases the risk of tBPAR one year after liver transplant, but everolimus administered without CNIs increases tBPAR.
-
Therapeutic drug monitoring for everolimus in kidney transplantation using 12-month exposure, efficacy, and safety data
Marc I Lorber,Claudio Ponticelli,John Whelchel,Hartmut W Mayer,John Kovarik,Yulan Li,Heinz Schmidli
DOI: https://doi.org/10.1111/j.1399-0012.2005.00326.x
Abstract:The aims of the current study were to determine whether therapeutic drug monitoring (TDM) might benefit kidney transplant recipients receiving everolimus, and to establish dosage recommendations when everolimus is used in combination with cyclosporine and corticosteroids. The analysis was based on data from 779 patients enrolled in two 12-month trials. Everolimus trough concentrations >/=3 ng/mL were associated with a reduced incidence in biopsy-proven acute rejection (BPAR) in the first month (p = 0.0001) and the first 6 months (p = 0.0001), and reduced graft loss compared with lower concentrations (4% vs. 20%, respectively). By contrast, cyclosporine in the standard concentration range had no impact on BPAR within the same timeframes. Most patients receiving everolimus 1.5 or 3 mg/d achieved trough concentrations above the therapeutic threshold of 3 ng/mL, regardless of reductions in cyclosporine dose. TDM simulation showed that just two dose adjustments would achieve median everolimus trough values >/=3 ng/mL in 95% of patients during the first 6 months. This investigation indicates that improved efficacy is likely when TDM is considered as an integral component of the immunosuppressive strategy of everolimus.
-
Safety, Tolerability, and Efficacy of Everolimus in De Novo Liver Transplant Recipients: 12- and 36-Month Results.
Gary Levy,Heinz Schmidli,Jeffrey Punch,Elizabeth Tuttle-Newhall,David Mayer,Peter Neuhaus,Didier Samuel,Bjorn Nashan,Juergen Klempnauer,Alan Langnas,Yvon Calmus,Xavier Rogiers,Michael Abecassis,Richard Freeman,Maarten Sloof,John Roberts,Lutz Fischer
DOI: https://doi.org/10.1002/lt.20707
2006-01-01
Liver Transplantation
Abstract:Everolimus is a macrolide immunosuppressive agent with known consistent absorption. In this double-blind study, we examined the safety and tolerability of everolimus vs. placebo in de novo liver transplant recipients. One hundred and nineteen liver allograft recipients were randomized to 1 of 4 groups: everolimus 0.5 mg bid, everolimus 1.0 mg bid, everolimus 2 mg bid, or placebo. Patients received oral cyclosporine to achieve a target trough level of 150-400 ng/mL in combination with prednisone. Primary and secondary endpoints of safety, tolerability, and efficacy were determined at 12 months, and patients were followed through 36 months. There was a trend toward fewer treated acute rejections in the everolimus group than in the placebo group: everolimus 0.5 mg: 39.3%; everolimus 1.0 mg: 30.0%; everolimus 2 mg: 29.0%; placebo: 40.0% (P = not significant). Adverse events were higher in everolimus-treated patients especially at the 4-mg/day dose, but there was no difference in the incidence of thrombocytopenia or leukopenia between all groups and renal function as determined by serum creatinine, and creatinine clearance remained stable to 36 months in everolimus-treated patients. Mean cholesterol and triglycerides increased from baseline in all treatment groups, and maximum levels were seen at 6 months. In conclusion, this study demonstrates that everolimus in combination with oral cyclosporine had an acceptable safety and tolerability profile, paving the way for additional studies in this transplant indication.
-
Early Initiation of Everolimus after Liver Transplantation: A Single-Center Experience.
Uta Herden,Antonio Galante,Lutz Fischer,Sven Pischke,Jun Li,Eike Achilles,Martina Koch,Bjoern Nashan,Martina Sterneck
DOI: https://doi.org/10.12659/aot.895800
2016-01-01
Annals of Transplantation
Abstract:BACKGROUND Evidence relating to early everolimus use after liver transplantation remains limited. MATERIAL AND METHODS Ninety-one adult patients undergoing liver transplantation at our center during 2007-2012 in whom everolimus therapy was initiated <3 months post-transplant were analyzed retrospectively. Everolimus was started on days 1-5 in 50 patients (group 1) and after day 5 in 41 patients (group 2). Most patients continued to receive low-dose cyclosporine (59.3%, target 50-80 ng/ml) or low-dose tacrolimus (25.3%; target 3-5 ng/ml). Mean follow-up was 4.6 years. RESULTS One-, three- and five-year patient survival rates were 80.5%, 74.2%, and 70.5%, respectively, and did not differ between groups 1 and 2. Six patients (6.6%) developed biopsy-proven acute rejection after a median of 47 days (range 27-356 days). Everolimus was discontinued due to adverse events in 21 patients (23.1%). Incisional hernia repair occurred in 14.0% and 9.4% of patients in group 1 and 2, respectively. Renal function remained stable during follow-up, despite poor baseline function. CONCLUSIONS Everolimus with very low-dose calcineurin inhibitor given immediately after liver transplantation appears safe and effective, achieving a low rejection rate with well-preserved renal function.
-
Evaluating the Efficacy, Safety and Evolution of Renal Function with Early Initiation of Everolimus-Facilitated Tacrolimus Reduction in De Novo Liver Transplant Recipients: Study Protocol for a Randomized Controlled Trial
Bjorn Nashan,Peter Schemmer,Felix Braun,Markus Dworak,Peter Wimmer,Hans Schlitt
DOI: https://doi.org/10.1186/s13063-015-0626-0
IF: 2.728
2015-01-01
Trials
Abstract:Introduction of calcineurin inhibitors had led to improved survival rates in liver transplant recipients. However, long-term use of calcineurin inhibitors is associated with a higher risk of chronic renal failure, neurotoxicity, de novo malignancies, recurrence of hepatitis C viral (HCV) infection and hepatocellular carcinoma. Several studies have shown that everolimus has the potential to provide protection against viral replication, malignancy, and progression of fibrosis, as well as preventing nephrotoxicity by facilitating calcineurin inhibitor reduction without compromising efficacy. The Hephaistos study evaluates the beneficial effects of early initiation of everolimus in de novo liver transplant recipients.
-
Everolimus Based Calcineurin Inhibitors Minimization Regimens in Renal Transplantation Recipients: A Meta-Analysis.
Z. Guo,F. Li,D. Wang,W. Ju,X. He
DOI: https://doi.org/10.1097/00007890-201407151-01804
2014-01-01
Transplantation
Abstract:Background Calcineurin inhibitors (CNIs) are widely-used immunosuppressants in renal transplantation, although it has been proved that CNIs can cause nephrotoxicity. Minimization of CNIs seems a better immunosuppression protocol after renal transplantation. Here we conduct a meta-analysis on the outcomes of CNI minimization by Everolimus in renal transplant recipients. Methods We included randomized trials with no year and language restriction. All data were analyzed using fix or random effect model. The primary endpoints were estimated glomerular filtration rate (eGFR), biopsy proven acute rejection (BPAR) and patient survival at month 12. The secondary endpoints were incidence of infection, blood creatinine level, allograft dysfunction, and other side effects such as hypertriglyceridemia. ResultseGFR was significantly improved in Everolimus group (MD=5.41, 95% CI 0.63 to 10.19, Z=2.22, P=0.03). There was no significant difference in BPAR episodes (RR=1.29, 95% CI=0.82 to 2.04, Z=1.10, P=0.27), patient survival (RR=0.88, 95% CI=0.45 to 1.75, Z=0.35, P=0.73), overall infection rate (RR=0.99, 95%CI=0.93 to 1.06, Z=0.28, P=0.78) and incidence of hypertriglyceridemia (RR=0.94, 95%CI=0.56 to 1.57, Z=0.24, P=0.81).Table: No Caption available.Conclusion The findings of this meta-analysis suggest that CNI minimization regimen with Everolimus elicits benefits in renal function, while not comprising immunosuppressive effects.
-
Conversion to everolimus in liver transplant patients with renal dysfunction
T Pérez,R Segovia,L Castro,J P Roblero,R Estela
DOI: https://doi.org/10.1016/j.transproceed.2011.06.009
Abstract:Calcineurin inhibitor (CNI) immunosuppressive therapy post-liver transplantation (OLT) is important to reduce graft rejection episodes. However, these drugs show important side effects, particularly renal dysfunction (RDF). Changing from CNI to a nonnephrotoxic drug, as mammalian target of rapamycin (mTOR) inhibitor may solve the problem. Our objective was to evaluate renal function (RF) among liver transplant patients initially receiving CNI, among whom the patients with RDF were converted completely or partially to an mTOR inhibitor like everolimus (EVE). We performed a prospective study in liver transplant patients from 2000 to 2009. Creatinine levels and creatinine clearances (Cockroft-Gault) expressed as mean values ± standard deviations were measured pre- and postswitch for comparisons using Wilcoxon nonparametric tests. Six patients were converted fully or partially to EVE. Their mean age at the moment of introducing the new therapy was 52.2 ± 13.6 years (range = 28-60). Immunosuppression time prior to switching from CNI to EVE was 23.8 ± 26.6 months (range = 6-70). Postconversion follow-up was 25.8 ± 16.5 months (range = 8-42). All patients showed improvement in RF. The creatinine level improvement was significant (P = .03) namely, from a mean of 2.26 ± 0.49 to 1.21 ± 0.57 mg/dL. Glomerular filtration rate improved from a mean of 40 ± 15.13 to 72.60 ± 17.3 mL/min/m(2) (P = .03). Conversion from CNI to EVE improved creatinine concentrations and creatinine clearances with long-term effects free of graft rejection.
-
Effect of everolimus on the glucose metabolic pathway in mouse skeletal muscle cells (C2C12)
Kayoko Yoshida,Chiyo K. Imamura,Kanako Hara,Mayumi Mochizuki,Yusuke Tanigawara
DOI: https://doi.org/10.1007/s11306-017-1236-5
2017-07-07
Metabolomics
Abstract:IntroductionEverolimus selectively inhibits mammalian target of rapamycin complex 1 (mTORC1) and exerts an antineoplastic effect. Metabolic disturbance has emerged as a common and unique side effect of everolimus.ObjectivesWe used targeted metabolomic analysis to investigate the effects of everolimus on the intracellular glycometabolic pathway.MethodsMouse skeletal muscle cells (C2C12) were exposed to everolimus for 48 h, and changes in intracellular metabolites were determined by capillary electrophoresis time-of-flight mass spectrometry. mRNA abundance, protein expression and activity were measured for enzymes involved in glycometabolism and related pathways.ResultsBoth extracellular and intracellular glucose levels increased with exposure to everolimus. Most intracellular glycometabolites were decreased by everolimus, including those involved in glycolysis and the pentose phosphate pathway, whereas no changes were observed in the tricarboxylic acid cycle. Everolimus suppressed mRNA expression of enzymes related to glycolysis, downstream of mTOR signaling enzymes and adenosine 5′-monophosphate protein kinases. The activity of key enzymes involved in glycolysis and the pentose phosphate pathway were decreased by everolimus. These results show that everolimus impairs glucose utilization in intracellular metabolism.ConclusionsThe present metabolomic analysis indicates that everolimus impairs glucose metabolism in muscle cells by lowering the activities of glycolysis and the pentose phosphate pathway.
endocrinology & metabolism
-
Suppression of Cytomegalovirus-Specific Cd8(+) T Cells by Everolimus
Nan Jin,Georg Malcherek,Jiju Mani,Romy Zurleit,Anita Schmitt,Baoan Chen,Mathias Freund,Anthony D. Ho,Michael Schmitt
DOI: https://doi.org/10.3109/10428194.2013.822496
2014-01-01
Leukemia & Lymphoma
Abstract:Abstract Everolimus (RAD-001) has recently been used as an immunosuppressive drug to treat patients after hematopoietic stem cell transplant (HSCT), thereby reducing cyclosporine-related nephrotoxicity. We studied the immunomodulatory effect of everolimus on mitogen-stimulated and particularly cytomegalovirus (CMV)-specific cytotoxic T cells. Proliferation of CD4+ and CD8+ T cells stimulated with staphylococcal endotoxin B and phytohemagglutinin was strongly inhibited at very low doses. Proliferation of CMV-specific CD8+ T cells could be completely suppressed. Similarly, the frequency of CMV-specific, cytokine-secreting and CD137-expressing CD8+ T cells decreased in a dose-dependent manner. However, interferon-γ (IFN-γ) secretion by CMV-specific CD8+ T cells remained unchanged, as could be demonstrated by intracellular cytokine staining. As reactivation of CMV plays a pivotal role in the outcome of patients after HSCT, attention must be paid to early detection and preemptive treatment of CMV reactivity in patients treated with everolimus.
-
Everolimus Mitigates the Risk of Hepatocellular Carcinoma Recurrence after Liver Transplantation
Paolo De Simone,Arianna Precisi,Quirino Lai,Juri Ducci,Daniela Campani,Piero Marchetti,Stefano Gitto
DOI: https://doi.org/10.3390/cancers16071243
2024-03-22
Cancers
Abstract:To obtain long-term data on the use of everolimus in patients who underwent liver transplantation for hepatocellular carcinoma, we conducted a retrospective, single-center analysis of adult recipients transplanted between 2013 and 2021. Patients on everolimus-incorporating immunosuppression were matched with those on tacrolimus using an inverse probability of treatment weighting methodology. Two propensity-matched groups of patients were thus compared: 233 (45.6%) receiving everolimus versus 278 (54.4%) on tacrolimus. At a median (interquartile range) follow-up of 4.4 (3.8) years after transplantation, everolimus patients showed a reduced risk of recurrence versus tacrolimus (7.7% versus 16.9%; RR = 0.45; p = 0.002). At multivariable analysis, microvascular infiltration (HR = 1.22; p < 0.04) and a higher tumor grading (HR = 1.27; p < 0.04) were associated with higher recurrence rate while being within Milan criteria at transplant (HR = 0.56; p < 0.001), a successful pre-transplant downstaging (HR = 0.63; p = 0.01) and use of everolimus (HR = 0.46; p < 0.001) had a positive impact on the risk of post-transplant recurrence. EVR patients with earlier drug introduction (≤30 days; p < 0.001), longer treatment duration (p < 0.001), and higher drug exposure (≥5.9 ng/mL; p < 0.001) showed lower recurrence rates versus TAC. Based on our experience, everolimus provides a reduction in the relative risk of hepatocellular carcinoma recurrence, especially for advanced-stage patients and those with earlier drug administration, higher drug exposure, and longer time on treatment. These data advocate for early everolimus introduction after liver transplantation to reduce the attrition rate consequent to chronic immunosuppression.
oncology
-
Induction Therapy and Mtor Inhibition: Minimizing Calcineurin Inhibitor Exposure in De Novo Renal Transplant Patients
Bjoern Nashan
DOI: https://doi.org/10.1111/ctr.12156
2013-01-01
Abstract:Use of induction therapy with mTOR inhibitor maintenance immunosuppression to facilitate reduced calcineurin inhibitor (CNI) exposure in de novo kidney transplant patients has been explored in a series of randomized trials. These studies have typically employed interleukin-2 receptor antagonist (IL-2RA) induction, in low or standard immunological risk recipients. Although no study has directly compared mTOR inhibition plus reduced CNI exposure with or without induction, inclusion of IL-2RA induction appears to permit a substantial reduction in CNI exposure without the need for high mTOR inhibitor dosing. IL-2RA induction with an mTOR inhibitor and steroids has consistently shown similar efficacy to standard-exposure CNI with mycophenolic acid and steroids and may improve renal function among patients who remain on the mTOR inhibitor-based regimen. With modern mTOR inhibitor dosing, wound healing complications are of less concern and may be no more frequent than in mycophenolic acid-based regimens. The incidence of cytomegalovirus infection appears lower in patients receiving de novo mTOR inhibition. The available evidence demonstrates that IL-2RA induction with an mTOR inhibitor can successfully reduce CNI exposure by at least half without a penalty in terms of rejection in low- or moderate-risk de novo transplant recipients and may offer renal and antiviral benefits.
-
Use of sirolimus in solid organ transplantation
Joshua J Augustine,Kenneth A Bodziak,Donald E Hricik
DOI: https://doi.org/10.2165/00003495-200767030-00004
Drugs
Abstract:Sirolimus is a mammalian target of rapamycin (mTOR) inhibitor that inhibits cell cycle progression and has proven to be a potent immunosuppressive agent for use in solid organ transplant recipients. The drug was initially studied as an adjunct to ciclosporin (cyclosporine) to prevent acute rejection in kidney transplant recipients. Subsequent studies have shown efficacy when combined with a variety of other immunosuppressive agents. The most common adverse effects of sirolimus are hyperlipidaemia and myelosuppression. The drug has unique antiatherogenic and antineoplastic properties, and may promote immunological tolerance and reduce the incidence of chronic allograft nephropathy. Although sirolimus is relatively non-nephrotoxic when administered as monotherapy, it pharmacodynamically enhances the toxicity of calcineurin inhibitors. Ironically, the drug has been used to facilitate calcineurin inhibitor-free protocols designed to preserve renal function after solid organ transplantation. Whether sirolimus can be used safely over the long term with low doses of calcineurin inhibitors requires further study. The use of sirolimus as a corticosteroid-sparing agent also remains to be proven in controlled trials. Postmarketing studies have revealed a number of unforeseen adverse effects including impaired wound healing and possibly proteinuria, oedema, pneumonitis and thrombotic microangiopathy. Overall, sirolimus is a powerful agent when used judiciously with other available immunosuppressants. As is true for all immunosuppressive drugs available for treatment of solid organ transplant recipients, the efficacy of the drug must be balanced against its considerable adverse effects.
-
Everolimus And Sirolimus Hypersensitivity Pneumonitis With Full Recovery: A Case Series
Michelle Dimza,Yomary Jimenez,Mohammad Al-Ani,Alex Parker,Mustafa Ahmed,Juan Aranda,Juan Vilaro
DOI: https://doi.org/10.1016/j.cardfail.2023.10.265
IF: 6.592
2024-01-01
Journal of Cardiac Failure
Abstract:Introduction Everolimus and sirolimus are immunosuppressants that inhibit mechanistic target of rapamycin (mTOR) used to prevent graft rejection after heart transplantation. Hypersensitivity pneumonitis is a known class effect toxicity of mTOR inhibitors. Prognosis is favorable if medication is stopped before there is extensive damage. We report two cases of hypersensitivity pneumonitis in patients receiving mTOR inhibitors. Case 1 A 45-year-old female who was 7 years S/P heart transplant for NICM presented to clinic for progressive shortness of breath for one month. Her post-transplant course was complicated by antibody-mediated rejection and allograft vasculopathy, for which mycophenolate mofetil (MMF) was substituted for everolimus 3months prior to her presentation. CT scan showed new ground glass opacities (GGO) throughout the right lung and the left lower lobe (figure 1a). An extensive infectious workup was negative. Bronchoscopy with BAL resulted in elevated polymorphonuclear leukocytes (55%), lymphocytes (4%), and macrophages (41%) with negative cultures and antigen testing. She remained hemodynamically stable on room air and was discharged on MMF instead of everolimus. After 2 months, her symptoms had improved, and repeat CT scan showed near complete resolution of prior infiltrates (Figure 1b). Case 2 A 41-year-old female who was 9 years S/P heart transplant for peripartum cardiomyopathy presented with 6 months of progressive shortness of breath and productive cough. MMF was switched to sirolimus for de novo donor-specific antibodies 6 months prior. On presentation, she was hemodynamically stable on room air. Labs were significant for WBC 20 x109/L with 48% eosinophils. CT scan showed patchy diffuse GGO (figure 2a). An extensive infectious workup was negative. Bronchoscopy with BAL showed 93% Eosinophils. The diagnosis of mTOR-induced eosinophilic pneumonitis was made, and sirolimus was stopped. She was discharged on a higher dose of MMF. Her symptoms continued to improve. Four months later, she had a repeat CT scan which showed resolution of GGO (figure 2b). Conclusion We present two cases of reversible mTOR inhibitor-induced hypersensitivity pneumonitis with full clinical and radiographic recovery with discontinuation of therapy. It is vital to screen for symptoms and recognize the diagnosis early as discontinuation of therapy can prevent significant morbidity. .
cardiac & cardiovascular systems
-
A comprehensive review of everolimus clinical reports: a new mammalian target of rapamycin inhibitor
Cheryle Gurk-Turner,Wana Manitpisitkul,Matthew Cooper
DOI: https://doi.org/10.1097/TP.0b013e31825b411c
2012-10-15
Transplantation
Abstract:As new immunosuppressive agents are introduced to the market, clinicians are faced with the daunting task of sifting through the published literature to decide the value that the agent will add to their own practice. We often must extrapolate information provided through study in other solid-organ transplantation populations than our specific area of interest as we interpret the results and outcomes. With these challenges in mind, this compilation of published work for the newest mammalian target of rapamycin inhibitor everolimus (Certican; Novartis Pharmaceuticals, Hanover, NJ) (Zortress; Novartis Pharmaceuticals, Basel, Switzerland) is intended to provide a concise but thorough presentation of available literature so that the reader who may be unfamiliar with the agent can make their own judgment. Both Ovid and PubMed search engines were queried with a particular focus on high-impact articles noted in the Web of Science or Citation Index. Work described solely in abstract or case report form was excluded, as well as meta-analyses or those that were editorial or commentary in nature. Included were publications presented using the English language that described adult human subjects who received a heart, lung, kidney, or liver allograft. The goal of this strategy was to allow for the inclusion of pertinent literature in an unbiased fashion. Tables are provided that outline trial specific information, leaving a discussion of major outcomes to the text of the review.
-
Everolimus Reduces Cancer Incidence and Improves Patient and Graft Survival Rates after Kidney Transplantation: A Multi-Center Study
Ryoichi Imamura,Ryo Tanaka,Ayumu Taniguchi,Shigeaki Nakazawa,Taigo Kato,Kazuaki Yamanaka,Tomoko Namba-Hamano,Yoichi Kakuta,Toyofumi Abe,Koichi Tsutahara,Tetsuya Takao,Hidefumi Kishikawa,Norio Nonomura
DOI: https://doi.org/10.3390/jcm11010249
IF: 3.9
2022-01-04
Journal of Clinical Medicine
Abstract:Kidney transplantation can prevent renal failure and associated complications in patients with end-stage renal disease. Despite the good quality of life, de novo cancers after kidney transplantation are a major complication impacting survival and there is an urgent need to establish immunosuppressive protocols to prevent de novo cancers. We conducted a multi-center retrospective study of 2002 patients who underwent kidney transplantation between 1965 and 2020 to examine patient and graft survival rates and cumulative cancer incidence in the following groups categorized based on specific induction immunosuppressive therapies: group 1, antiproliferative agents and steroids; group 2, calcineurin inhibitors (CNIs), antiproliferative agents and steroids; group 3, CNIs, mycophenolate mofetil, and steroids; and group 4, mammalian target of rapamycin inhibitors including everolimus, CNIs, mycophenolate mofetil, and steroids. The patient and graft survival rates were significantly higher in groups 3 and 4. The cumulative cancer incidence rate significantly increased with the use of more potent immunosuppressants, and the time to develop cancer was shorter. Only one patient in group 4 developed de novo cancer. Potent immunosuppressants might improve graft survival rate while inducing de novo cancer after kidney transplantation. Our data also suggest that everolimus might suppress cancer development after kidney transplantation.
medicine, general & internal
-
Phosphorylation of mTOR and S6RP predicts the efficacy of everolimus in patients with metastatic renal cell carcinoma
Siming Li,Yan Kong,Lu Si,Zhihong Chi,Chuanliang Cui,Xinan Sheng,Jun Guo
DOI: https://doi.org/10.1186/1471-2407-14-376
IF: 4.638
2014-01-01
BMC Cancer
Abstract:Background The incidence of renal cell cancer (RCC) has been increasing for the past decade, and the 5-year survival for patients with metastatic RCC (mRCC) is rather low. Everolimus (RAD001), a new inhibitor for mammalian target of rapamycin (mTOR), is generally well tolerated, and demonstrates clinical benefit to patients with anti-VEGF-refractory mRCC. However, factors for selection of patients who may benefit from everolimus remain largely unknown. Here we aimed to explore potential molecular indicators for mRCC patients who may benefit from everolimus treatment. Methods Paraffin-embedded tumor tissue specimens derived from 18 mRCC patients before everolimus treatment, who participated the phase 1b trial of everolimus in VEGF receptor (VEGFR)-tyrosine kinase inhibitor (TKI)-refractory Chinese patients with mRCC (clinicaltrials.gov, NCT01152801), were examined for the expression levels of phosphorylated AKT, mTOR, eukaryotic initiation factor 4E (eIF4E) binding protein-1 (4EBP1) and 40S ribosomal protein S6 (S6RP) by immunohistochemistry. Clinical benefit rate (complete response [CR], partial response [PR], plus stable disease [SD] ≥ 6 months) and progression-free survival time (PFS) were correlated with expression levels of these mTOR-associated molecules. Results In these 18 patients, there were 1 PR, 15 SDs (including 9 SDs ≥ 6 months), and 2 progressive diseases (PD). The clinical benefit rate (CBR) was 55.6% (10/18), and the median PFS time was 8.4 months. Patients with positive expression of phospho-mTOR showed a better CBR (71.4% versus 0%, P = 0.023) and PFS time (11.3 versus 3.7 months, P = 0.001) than those patients with negative expression. The median PFS of patients with positive phospho-S6RP expression was longer (11.3 versus 3.7 months, P = 0.002) than that of patients negative for phospho-S6RP expression. However, expression levels of phospho-4EBP1 and phospho-AKT were unassociated to efficacy of everolimus treatment with respect to CBR and PFS. Co-expression of phosphorylated mTOR, S6RP and/or 4EBP1 may improve the predictive value of the biomarkers for patients treated with everolimus. Conclusions The expression levels of phospho-mTOR and phospho-S6RP may be potential predictive biomarkers for efficacy of everolimus in patients with mRCC. Combining examinations of phosphorylated mTOR, S6RP and/or 4EBP1 may be a potential strategy to select mRCC patients sensitive to mTOR inhibitor treatment.