Review Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Transplant. Dec 24, 2013; 3(4): 68-77
Published online Dec 24, 2013. doi: 10.5500/wjt.v3.i4.68
Novel immunosuppressive agents in kidney transplantation
Karen L Hardinger, Department of Pharmacy Practice and Administration, University of Missouri-Kansas City, Kansas City, MO 64108, United States
Daniel C Brennan, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States
Author contributions: All authors have made substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data, drafting the article or revising it critically for important intellectual content, and final approval of the version to be published.
Correspondence to: Karen L Hardinger, PharmD, BCPS, Clinical Associate Professor of Pharmacy Practice, Department of Pharmacy Practice and Administration, University of Missouri-Kansas City, 2464 Charlotte Street, Rm 2241, Kansas City, MO 64108, United States. hardingerk@umkc.edu
Telephone: +1-816-2769023 Fax: +1-816-2764751
Received: June 28, 2013
Revised: August 26, 2013
Accepted: October 16, 2013
Published online: December 24, 2013

Abstract

Excellent outcomes have been achieved in the field of renal transplantation. A significant reduction in acute rejection has been attained at many renal transplant centers using contemporary immunosuppressive, consisting of an induction agent, a calcineurin inhibitor, an antiproliferative agent plus or minus a corticosteroid. Despite improvements with these regimens, chronic allograft injury and adverse events still persist. The perfect immunosuppressive regimen would limit or eliminate calcineurin inhibitors and/or corticosteroid toxicity while providing enhanced allograft outcomes. Potential improvements to the calcineurin inhibitor class include a prolonged release tacrolimus formulation and voclosporin, a cyclosporine analog. Belatacept has shown promise as an agent to replace calcineurin inhibitors. A novel, fully-human anti-CD40 monoclonal antibody, ASKP1240, is currently enrolling patients in phase 2 trials with calcineurin minimization and avoidance regimens. Another future goal of transplant immunosuppression is effective and safe treatment of allograft rejection. Novel treatments for antibody mediated rejection include bortezomib and eculizumab. Several investigational agents are no longer being pursed in transplantation including the induction agents, efalizumab and alefacept, and maintenance agents, sotrastaurin and tofacitinib. The purpose of this review is to consolidate the published evidence of the effectiveness and safety of investigational immunosuppressive agents in renal transplant recipients.

Key Words: Review, Immunosuppression, Investigational agents, Renal/kidney transplant

Core tip: Many new agents are being studied that may improve outcomes after renal transplantation. Potential improvements to the calcineurin inhibitor class include a recently Food and Drug Administration approved, prolonged release tacrolimus formulation and voclosporin, a cyclosporine analog. A novel, fully-human anti-CD40 monoclonal antibody, ASKP1240, is currently enrolling patients in phase 2 trials with calcineurin minimization and avoidance regimens. Novel treatments for antibody mediated rejection include bortezomib and eculizumab.



INTRODUCTION

Although significant advances in renal transplant immunosuppression have occurred in the past decades, a vital need to improve long-term survival remains. Currently, immunologic causes of early graft failure have become rare, yet late graft loss has remained virtually unchanged. One of the most common causes for graft loss is chronic allograft nephropathy. Additionally, significant drug improvements in transplantation have come with the expense of side effects. Many of these adverse events, including new onset diabetes after transplant, dyslipidemia, and hypertension, may contribute to cardiovascular related deaths after transplantation. The ideal immunosuppressive regimen would improve long-term outcomes while minimizing exposure to drug toxicity and infection.

Induction agents are typically antibodies (anti-thymocyte globulins) or interleukin 2 receptor antagonists (basiliximab). Another induction agent, alemtuzumab has been removed from the United States market, but is still available through a special manufacturer program. The five drug classes that currently comprise maintenance regimens may include calcineurin inhibitors (cyclosporine and tacrolimus), mTOR inhibitors (sirolimus and everolimus), antiproliferative agents (azathioprine and mycophenolic acid), costimulation blockers (belatacept) and corticosteroids (Table 1). KDIGO Clinical Practice Guidelines suggest that first-line agents should include basiliximab induction for low-risk patients and an anti-thymocyte globulin for high-risk patients in conjunction with maintenance immunosuppression including tacrolimus and mycophenolate[1]. Potential improvements to the calcineurin inhibitor class include a prolonged release tacrolimus formulation and voclosporin, a cyclosporine analog (Table 2). A novel, anti CD-40 molecule has completed phase 1 studies.

Table 1 Summary of investigational and available agents.
GenericBrandFDA indicationCompany
Alefacept12AmeviveTreatment of moderate-to-severe chronic plaque psoriasis in adults who are candidates for systemic therapy or phototherapyAstellas
Alemtuzumab2CampathTreatment of B-CLLBerlex Laboratories
ASKP1240Not FDA approvedAstellas
AzathioprineImuranAdjunctive therapy in prevention of rejection of kidney transplants; management of active rheumatoid arthritisGeneric manufacturers
BasiliximabSimulectPrevention of acute rejection in kidney transplantationNovartis
BelataceptNulojixPrevention of acute rejection in renal transplant recipientsBristol-Myers-Squibb
BortezomibVelcadeTreatment of multiple myeloma; treatment of relapsed or refractory mantle cell lymphomaMillenium Pharmaceuticals
CyclosporineNeoralPrevention of acute rejection in renal transplant recipientsNovartis
EculizumabSolirisTreatment of PNH to reduce hemolysis and aHUSAlexion Pharmaceuticals
Efalizumab12RaptivaManagement of moderate to severe chronic plaque psoriasis in adultsGenentech
EverolimusAfinitor, ZortressTreatment of advanced renal cell cancer (Afinitor®); treatment of subependymal giant cell astrocytom associated with tuberous sclerosis (Afinitor®); treatment of advanced, metastatic or unresectable pancreatic neuroendocrine tumors (Afinitor®); prophylaxis of organ rejection in patients at low-moderate immunologic risk receiving renal transplants (Zortress®)Novartis
Mycophenolate MofetilCellceptProphylaxis of organ rejection concomitantly with cyclosporine and corticosteroids in patients receiving allogeneic renal cardiac, or hepatic transplantsGenentech
Mycophenolate SodiumMyforticProphylaxis of organ rejection concomitantly with cyclosporine and corticosteroids in patients receiving allogeneic renal transplantationNovartis
Horse or Rabbit anti-thymocyte GlobulinAtgam or ThymoglobulinTreatment of corticosteroid resistant rejection in kidney transplantationPfizer/Sanofi
RituximabRituxanTreatment of CD20-positive non-Hodgkin’s lymphomas ; Treatment of moderately- to severely-active rheumatoid arthritis in adult patients with inadequate response to one or more TNF antagonists; Treatment of Wegener’s granulomatosis; Treatment of microscopic polyangiitisGenentech
SirolimusRapamunePrevention of acute rejection in renal transplant recipientsPfizer
Sotrastaurin, AEB-0711Not FDA approvedNovartis
TacrolimusPrografPrevention of acute rejection in renal transplant recipientsAstellas
Tacrolimus Prolonged ReleaseAstragraf XLPreventing organ rejection in kidney transplant recipients, as combination therapy with mycophenolate mofetil and corticosteroids, with or without tasiliximab inductionAstellas
Tolfacitinib1XeljanzTreatment of moderate to severe rheumatoid arthritisPfizer
VoclosporinNot FDA approvedIsotechnika Pharma
Table 2 Non-Food and Drug Administration approved/investigational agents and their mechanism.
Drug nameMechanism of action
Induction
Efalizumab1Humanized antibody, CD11a/LFA-1
Alefacept1Costimulation inhibitor, CD2 LFA3
Maintenance
Voclosporin, ISA247Calcineurin inhibitor
Sotrastaurin, AEB0711Protein kinase C inhibitor
Tofacitinib, CP-6905501JAK 3 inhibitor
ASKP1240Anti-CD40 monoclonal antibody
Treatment of Antibody Medicated Rejection
BortezomibProteasome inhibitor
EculizumabMonoclonal antibody, C5 complement protein

Another area for improvement is treatment of humoral rejection. Historically, treatment has been difficult and not well studied. Humoral rejection is typically treated with intravenous immunoglobulin, rituximab and plasmapheresis. Investigational treatments for antibody mediated rejection that will be discussed include bortezomib and eculizumab.

In the past years, several clinical trials have been unsuccessful and therefore many agents are no longer being pursued for transplantation. These agents include two inductions agents, efalizumab and alefacept, and two maintenance agents, sotrastaurin (a protein kinase C inhibitor) and tofacitinib (a JAK 3 inhibitor). This review article will update a previously published article found in this journal[2] on the effectiveness and safety of novel immunosuppressive agents.

PREVENTATIVE AGENTS
Alternatives to currently available calcineurin inhibitors

Calcineurin inhibitors have revolutionized post-transplantation immunosuppressive regimens by significantly lowering acute rejections rates. Yet, long-term use of these drugs has been associated with the development of chronic allograft nephropathy and adverse events. New immunosuppressive agents that eliminate these issues are needed. Prolonged release tacrolimus (Astragraf XL®, Astellas) has been approved for use in various European countries, Canada and the United States (in July 2013). The expectation is that the products once daily, rather than twice daily, dosing will improve adherence in transplant recipients. Large, randomized, phase 3 studies have compared prolonged release-tacrolimus compared to tacrolimus with similar efficacy and safety outcomes[3,4]. Of note, tacrolimus levels may be slightly lower with prolonged release tacrolimus compared to twice daily tacrolimus patients[5-7], although serum creatinine, creatinine clearance and estimated glomerular filtration rate were very similar. Prolonged release tacrolimus has a non-inferior efficacy profile with convenient daily dosing which is expected to improve patient compliance. Drug cost may influence the widespread use of this product as generic tacrolimus formulations are now available.

A novel calcineurin inhibitor, voclosporin (ISA 247, Isotechnika Pharma, Inc.) is being investigated in solid organ transplant, uveitis, and psoriasis[8-11]. Animal studies demonstrated that voclosporin, a cyclosporine analogue, had a higher affinity and greater in-vivo potency[12,13]. PROMISE, a phase 2b trial of low risk renal transplant recipients with immediate allograft function (n = 334) compared low (0.4 mg/kg), medium (0.6 mg/kg) and high (0.8 mg/kg) dose voclosporin to tacrolimus (0.05 mg/kg), in combination with a standard immunosuppressive regimen (anti-CD25 antibody, mycophenolate mofetil, and corticosteroids). Rejection rates were non-inferior to tacrolimus (11%, 9%, 2 %, and 6% respectively) and renal function was clinically similar (69-72 mL/min) at 6 mo after transplantation[8]. The incidence of new onset diabetes after transplantation was significantly lower in the low dose voclosporin group (1.6% vs 16.4% tacrolimus), but not in the medium (5.7%) and high dose (17.7%) arms[8]. The major limitation of this trial was that only low risk patients were studied. Low to medium dose voclosporin may provide adequate immunosuppression with a lower incidence of new onset diabetes after transplantation. A large, phase 3 (n = 598) trial is planned for 2013.

Recently, pharmacokinetic data of voclosporin was presented at the American Transplant Congress[14-17]. Researchers have learned that voclosporin should be given on an empty stomach and that dosage adjustment may be needed in severe renal failure (< 30 mL/min) and mild to moderate hepatic impairment[14-16]. Optimal trough concentrations should be targeted between 35-60 ng/mL[17].

Belatacept (Nulojix®, Bristol Myers Squibb) is a second generation co-stimulation blocker (CD80 antagonism) that received Food and Drug Administration (FDA) approval for use in kidney transplantation in June of 2011. Belatacept is contraindicated in patients that are Ebstein-Barr virus seronegative, because of high incidence of post-transplant lymphoproliferative seen in clinical trials[19-22]. Belatacept is administered as a well-tolerated intravenous infusion over 30 min. The recommended dosing is 10 mg/kg administered, prior to transplantation, on day 5, and at the end of weeks 2, 4, 8, and 12, then 5 mg/kg every 4 wk (plus or minus 3 d). The chronic intravenous administration could prove beneficial in increasing patient compliance with less frequent (monthly) infusions. In contrast, it may be perceived as a barrier to patients without social support that cannot readily access an infusion center. Administration and drug costs may also influence prescribing patterns and patient compliance.

Belatacept is the first immunosuppressive to demonstrate a renal benefit over a calcineurin inhibitor based regimen[18-22]. One limitation of the early belatacept trials (BENEFIT and BENEFIT-EXT) was that cyclosporine, a less contemporary immunosuppressive, was utilized[19-22]. In a phase 2, 1 year randomized study, belatacept/mycophenolate mofetil, belatacept/sirolimus and tacrolimus/mycophenolate mofetil, in combination with rabbit antithymocyte globulin and without corticosteroids were compared (n = 89)[23]. Acute rejection was highest in the belatacept/mycophenolate mofetil arm, graft loss was lowest in the tacrolimus/mycophenolate arm, and renal function was improved in the belatacept arms.

As an alternative to de novo immunosuppression, a conversion trial recently tested the hypothesis that belatacept-based regimens may provide a treatment option in patients already being treated with calcineurin-based maintenance immunosuppression[24]. Patients with stable graft function (calculated glomerular filtration rate between 35-75 mL/min) were randomized to either switch to belatacept (n =84) or continue calcineurin inhibitor treatment (n = 89). Despite a higher acute rejection rate in the belatacept group, the relative renal benefit of belatacept was observed in patients switched from either cyclosporine (+7.8 mL/min) or tacrolimus (+8.9 mL/min), and was observed regardless of baseline renal function. Patient survival, graft survival and the overall safety profile was similar between groups.

The impact of belatacept on long-term cardiovascular profiles is yet to be determined. An analysis of the pooled data from the BENEFIT AND BENEFIT-EXT trials showed lower blood pressures, lower non HDL cholesterol, lower triglycerides and less new onset diabetes mellitus after transplanation in the belatacept-treated patients versus the cyclosporine treated patients[25]. Yet, in a post-hoc analysis in patients with pre-existing diabetes from the BENEFIT and BENEFIT-EXT, 12 mo patient survival, graft survival, and renal function were similar between belatacept and cyclosporine treated patients[26]. Further trials are needed to explore the long-term outcomes, the impact of Epstein-Barr virus on post-transplant lymphoproliferative disease, and chronic allograft nephropathy. These trials should include contemporary immunosuppressive regimens.

A fully human anti-CD40 monoclonal antibody, ASKP1240 (Astellas®), has shown promise in phase 1 studies[27-29]. The first human, phase 1 study of healthy subjects (n = 12) demonstrated that the antibody was safe and well-tolerated[28]. Subsequently, a phase 1b trial, was performed in de novo kidney transplant recipients that received a single intravenous dose of 50 mg (n = 10), 100 mg (n = 9), 200 mg (n = 10), 500 mg (n = 9) or placebo (n = 8), no induction and standard maintenance immunosuppression per each center’s protocol[26]. ASKP1240 exhibited non-linear pharmacokinetics and was well tolerated at all doses. Acute rejection occurred in 3 patients in the 50 mg arm, 3 patients in the 500 mg arm and 1 patient in the placebo arm. The incidence of infection was not dose dependent. A phase 2 trial will compare the efficacy of ASKP1240 with calcineurin avoidance (basiliximab induction, ASKP1240, mycophenolate mofetil, and steroids) to the standard of care immunosuppressive regimen (basiliximab induction + tacrolimus + mycophenolate mofetil + steroids). In addition, the study will compare the efficacy of calcineurin inhibitor minimization-mycophenolate mofetil avoidance (basiliximab induction, ASKP1240, tacrolimus and steroids) to the standard of care immunosuppressive regimen.

TREATMENT OF ANTIBODY MEDIATED REJECTION

Antibody mediated rejection is an important cause of acute and chronic graft failure. Acute and chronic antibody mediated rejections are difficult to treat, because they are typically less responsive to conventional anti-rejection therapy. Treatment regimens for acute antibody mediated rejection may include one or more of the following: plasmapheresis, intravenous immunoglobulin (IVIG), and rituximab[30-38], although these regimens are not well-studied. A recent meta-analysis of over 10000 citations on treatment of antibody-mediated rejection concluded that data describing these treatments are of low or very low quality[34]. The first, prospective, randomized study comparing these strategies (plasmapheresis/IVIG/rituximab vs IVIG alone) demonstrated improved graft survival in the combination group[38]. Little guidance is given by the KDIGO Clinical Practice Guidelines, they suggest treating antibody-mediated acute rejection with one or more of the following alternatives with or without corticosteroids: plasma exchange; intravenous immunoglobulin; anti-CD20 antibody; lymphocyte-depleting antibody (Grade 2C Recommendation)[1]. Two investigational treatments for antibody mediated rejection include bortezomib and eculizumab.

Bortezomib (Velcade®, Millenium Pharmaceuticals) has been used for treatment of acute antibody mediated rejection, although it is approved for multiple myeloma in the United States (2010). It inhibits the degradation of cell-cycle regulatory proteins resulting in cell-cycle death via apoptosis. Bortezomib is metabolized via the cytochrome P450 system, a major substrate of 2C19 and 3A4 and inhibitor of 2C19, and therefore several drug interactions may occur including ketoconazole, clopidogrel, and grapefruit juice. Adverse events associated with bortezomib may include neutropenia, thrombocytopenia, nausea, vomiting, diarrhea, constipation (up to 50%), peripheral neuropathy (up to 30%), hypotension, QT prolongation, heart failure, pneumonitis and pneumonia. One case series of 52 transplant patients treated for antibody mediated rejection or desensitization reported bortezomib associated toxicity to be low, most commonly reported as manageable anemia or peripheral neuropathy[39]. Dosing of bortezomib is 1.3 mg/m2 on days 1, 4, 8, 11. No adjustments are necessary for renal impairment, but the dosage should be reduced by one-half for moderate to severe hepatic impairment.

Case series have reported the use of bortezomib to remove HLA antibodies in live-donor transplant recipients with HLA alloantibodies[40,41] and to treat antibody and cell-mediated acute rejection[42-51]. Few comparative trials have been performed. One German, historical control study of 10 bortezomib-treated patients (4 doses of 1.3 mg/m2) vs 9 rituximab-treated patients (one fixed dose of 500 mg) with antibody mediated rejection showed improved survival in the bortezomib treated group with an 18 mo graft survival of 60% vs 11% in the rituximab group[52]. All patients received plasmapheresis and intravenous immune globulin (30 g). Randomized trials are needed to determine the influence of bortezomib on antibody removal.

Eculizumab (Soliris®, Alexion Pharmaceuticals) is a humanized monoclonal IgG antibody that binds to complement protein C5 and blocks the activation of terminal complement. It is FDA approved for paroxysmal nocturnal hemoglobinuria and atypical hemolytic uremic syndrome. The most common side effects are headache, nausea, fatigue, back pain, cough and nasopharyngitis. Fatal immune hemolytic anemia following allogeneic stem cell transplantation has been reported[53]. Vaccination with meningococcal vaccine at least 2 wk prior to initiation of treatment with continued long-term prophylaxis is recommended. Eculizumab should not be used in patients with serious infections.

Case studies in renal transplant recipients have reported successful treatment of atypical hemolytic uremic syndrome and thrombotic microangiopathy with eculizumab[54-66]. Eculizumab has also been successful in reducing antibodies in a highly sensitized patient with positive crossmatches prior to live donor transplant[67] and in prevention of antibody mediated rejection in a case series of patients with donor specific antibodies and positive flow cytometry cross-matches (n = 4)[68]. In a larger case-control study, patients with donor specific antibodies who received pre-transplant plasmapheresis and post-transplant eculizumab were compared to historical controls[69]. At a median follow up of 12 mo for the eculizumab group, antibody mediated rejection occurred in 7.7 % (2/26) in the eculizumab group compared to 41% (21/51) in the control group (P < 0.01). One-year protocol biopsy revealed transplant glomerulopathy in 6.7% (1/15) eculizumab-treated recipients and in 35.7% (15/42) of control patients (P = 0.044). Eculizumab use has also been described in an ABO-incompatible deceased-donor kidney and pancreas transplant with a severe antibody-mediated rejection[70,71].

Eculizumab 600 mg weekly for six doses with plasmapheresis has also been successful in reversing refractory, early (mean time 6.5 d), acute antibody mediated rejection in four transplant recipients[72]. Mean follow up time is 6.4 ± 5.7 mo, and while antibodies persisted in the majority of the patients, the allografts are functioning and infectious complications have not occurred. Successful use of eculizumab has also been reported in two patients with antibody mediated rejection associated with thrombotic microangiopathy[73] and three patients with resistant antibody mediated rejection[74,75].

Despite the small sample size and lack of randomized controls, these studies are encouraging, and although larger studies and long-term follow-up are needed, bortezomib and eculizumab may play a major role in acute antibody mediated therapy in the future. Their role in transplant desensitization may be better elucidated as more clinical data and well-designed clinical trials become available. Current and future trials of bortezomib and eculizumab are listed in Table 3[76].

Table 3 Clinical trials.
AgentIdentifierStudy nameStart date
ASKP1240NCT01780844A Study to Assess the Efficacy and Safety of ASKP1240 in de Novo Kidney Transplant RecipientsFebruary 2013
VoclosporinNCT01586845Safety and Efficacy Study of Voclosporin and Tacrolimus in TransplantationMarch 2013
Prolonged Release TacrolimusNCT01294020Study to Compare the Pharmacokinetics of Tacrolimus in Stable Pediatric Allograft Recipients Converted From Prograf® to Advagraf®May 2011
BortezomibNCT01873157Bortezomib in Late Antibody-mediated Kidney Transplant Rejection (BORTEJECT)October 2013
NCT01349595Impact of Proteasome Inhibition on Anti-Donor HLA Antibody Production After Kidney TransplantationDecember 2011
NCT01842074Desensitization With Bortezomib Before a Living Kidney Donation (VELDON)January 2013
NCT01502267Desensitization Protocol for Highly Sensitized Patients on the Waiting List for Kidney TransplantJanuary 2010
NCT00722722The Impact of Velcade on Antibody Secreting Cells in Sensitized Renal Allograft CandidatesJune 2008
EculizumabNCT01349595Impact of Proteasome Inhibition on Anti-Donor HLA Antibody Production After Kidney TransplantationDecember 2011
NCT01327573Eculizumab Therapy for Chronic Complement-Mediated Injury in Kidney TransplantationMarch 2011
NCT01095887Eculizumab Added to Conventional Treatment in the Prevention of Antibody-mediated Rejection in ABO Blood Group Incompatible Living Donor Kidney Transplantation (ABOi)March 2010
NCT01403389A Study of the Activity of Eculizumab for Prevention of Delayed Graft Function In Deceased Donor Kidney TransplantAugust 2011
NCT01567085Safety and Efficacy Of Eculizumab In The Prevention Of Antibody Mediated Rejection (AMR) In Sensitized Recipients Of A Kidney Transplant From A Deceased DonorMay 2012
NCT01106027Dosing Regimen of Eculizumab Added to Conventional Treatment in Positive Crossmatch Deceased Donor Kidney TransplantMarch 2010
NCT01399593Safety and Efficacy of Eculizumab to Prevent AMR in Living Donor Kidney Transplant Recipients Requiring DesensitizationSeptember 2011
AGENTS NO LONGER BEING INVESTIGATED

Efalizumab (Raptiva®, Genentech) works an immunosuppressant by binding to the CD11a subunit of lymphocyte function-associated antigen 1 (LFA-1) and inhibiting white blood cell migration. This once weekly intramuscular injection was indicated for the treatment of chronic moderate-to-severe plaque psoriasis, but has been associated with an increased risk for progressive multifocal leukoencephalopathy (PML) and was withdrawn from the market in April of 2009[77]. Likewise, clinical trials in renal transplant recipients have not been successful due to higher rates of lymphoproliferative disease[78].

Alefacept (Amevive®, Astellas Pharmaceuticals) is a CD2-LFA3 co-stimulation inhibitor[79,80], was an intramuscular injection indicated for treatment of moderate-to-severe chronic plague psoriasis. Alefacept was voluntarily withdrawn from the market by Astellas Pharmaceuticals in December of 2011 due to “business needs”[81]. Prior to the discontinuation, alefacept was being developed for use in conjunction with tacrolimus, mycophenolate mofetil and steroids in renal transplantation. In a phase 2, de novo study of adult kidney transplant patients alefacept (vs placebo) resulted in similar survival and rejection rates, however the incidence of malignancy was higher in the alefacept arm[82].

Sotrastaurin (AEB071, Novartis), a protein kinase inhibitor, initially proved to have a good tolerability profile with few adverse effects[83]. Sotrastaurin development has been halted due high rejection rates (up to 40%) in de novo transplant recipients despite promising results with renal function and a low toxicity profile[83-87].

Tofacitinib (Xeljanz®, Tofacitinib CP-690550, Pfizer Inc.), is a kinase inhibitor with immunosuppressant properties that was FDA approved for moderate to severe rheumatoid arthritis in November of 2012. Tofacitinib is a small molecule agent which exhibits selective inhibition for the JAKs, thus inactivating the JAK/STAT dependent IL-2 induced T-cell proliferation.

Tofacitinib was being studied as a drug to be used in place of calcineurin inhibitors along with other anti-metabolite agents in two phase 2 clinical trials. In a small, initial, clinical study on de novo kidney allograft recipients comparing a tofacitinib regimen at 15 mg twice daily (CP15) and 30 mg twice daily (CP30) with tacrolimus, researchers reported the 6-mo biopsy-proven acute rejection rates to be 1 of 20, 4 of 20 and 1 of 21 for CP15, CP30 and tacrolimus groups respectively and concluded the 15 mg bid regimen to be similar to the tacrolimus regimen[88]. All patients received interleukin-2 receptor antagonist induction, mycophenolic acid and corticosteroids. In a subsequent, larger phase-2 trial (n = 331), a standard cyclosporine regimen was compared with a 15 mg twice daily regimen of tofacitinib which is subsequently switched to 10 mg twice daily after 3 mo (less-intensive) and another 15 mg twice daily regimen of tofacitinib which is switched to 10 mg twice daily after 6 mo (more-intensive)[89]. The biopsy proven acute rejection at 6 mo with the low-dose group (11%) was lower than the more-intensity or cyclosporine groups (7% and 9%, respectively). In terms of glomerular filtration rate at 12 mo, the tofacitinib groups (less-intensity: 65 mL/min and more-intensity: 65 mL/min) showed a significant difference in preservation of renal function compared to the cyclosporine group (54 mL/min). In this study, there was a lower incidence of chronic allograft nephropathy in the more intense and less intense groups (25% and 24% respectively) compared to the cyclosporine group (48%).

The smaller clinical study reported a high incidence of BK virus in the CP30 group (4/20) and a higher 6 mo rate of CMV disease (4/20) compared to CP15 and tacrolimus (2/20 and 0/20 respectively)[88]. Some other common abnormalities noted with this agent were trends towards higher lipid elevations, anemia and neutropenia during the first 6 mo of the treatment when the mycophenolate mofetil dose was high. In the larger, phase 2 trial, there were fewer cases of new-onset diabetes in the more-intense and less-intense groups (9.9% and 9.3% respectively) compared to cyclosporine (20.8%)[89]. The rate of serious infections, BK virus nephritis, post-transplant lymphoproliferative disorder and CMV disease was higher in the tofacitinib groups. The overall findings of the phase 2 studies suggest that tofacitinib is effective in preventing acute rejection and chronic allograft nephropathy, although this was achieved at the expense of hematological toxicity and over-immunosuppression when used in combination with mycophenolate mofetil. Although research has shown that safety may be improved by concentration-controlled dosing[90], tofacitinib development has been discontinued.

CONCLUSION

Induction agents are typically antibodies (anti-thymocyte globulins) or interleukin 2 receptor antagonists (basiliximab). Alemtuzumab has been removed from the United States market, but is available through the manufacturer through a special program. Many questions remain surrounding the use of potent induction agents including whether or not the use is associated with infection and malignancy, if the use is cost-effective, and if there is a true graft survival benefit. Due to poor clinical outcomes, induction investigational agents including, efalizumab and alefacept, are no longer being studied. Maintenance immunosuppressives may show some promise with future novel agents. Prolonged release tacrolimus provides once daily dosing of this product and hopefully will simplify a complex post-transplant immunosuppressive regimen. It is unknown if the perceived benefits will outweigh the cost of this product. Voclosporin, a cyclosporine analog, has not shown superior efficacy outcomes, but perhaps improvement in the safety profile (namely new-onset diabetes after transplant) will secure its place in transplant immunotherapy as the phase 3 trials are underway. ASKP, an anti-CD40 antibody, has successfully completed phase 1 studies and phase 2 trials are ongoing. Although belatacept has shown promise, two other investigational maintenance agents, sotrastaurin and tolfacitinib, will not be studied further in transplantation.

Treatment regimens for acute humoral rejection may include one or more of the following: plasmapheresis, intravenous immunoglobulin, and rituximab. Investigations of bortezomib and eculizumab have been hindered by small, non-randomized trials. Although results are encouraging, larger studies and long-term follow-up is ongoing.

At this point in time, there are very few immunosuppressants in clinical trials. Although some investigational agents have shown promise, tailoring available agents may need to be the short-term focus for transplant recipients. Hopefully, modifying exist regimens and approval of investigational agents will satisfy the ultimate goal of transplantation to improve long-term survival without toxicity or infection.

Footnotes

P- Reviewers: Shou FZ, Trimarchi H S- Editor: Song XX L- Editor: A E- Editor: Yan JL

References
1.  Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group. KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant. 2009;9 Suppl 3:S1-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 666]  [Cited by in F6Publishing: 987]  [Article Influence: 65.8]  [Reference Citation Analysis (0)]
2.  Kalluri HV, Hardinger KL. The current state of renal transplant immunosuppression: present and future. World J Transplant. 2012;2:51-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 51]  [Cited by in F6Publishing: 42]  [Article Influence: 3.5]  [Reference Citation Analysis (2)]
3.  Silva HT, Yang HC, Abouljoud M, Kuo PC, Wisemandle K, Bhattacharya P, Dhadda S, Holman J, Fitzsimmons W, First MR. One-year results with extended-release tacrolimus/MMF, tacrolimus/MMF and cyclosporine/MMF in de novo kidney transplant recipients. Am J Transplant. 2007;7:595-608.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 146]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
4.  Krämer BK, Charpentier B, Bäckman L, Silva HT, Mondragon-Ramirez G, Cassuto-Viguier E, Mourad G, Sola R, Rigotti P, Mirete JO. Tacrolimus once daily (ADVAGRAF) versus twice daily (PROGRAF) in de novo renal transplantation: a randomized phase III study. Am J Transplant. 2010;10:2632-2643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 132]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
5.  Jelassi ML, Lefeuvre S, Karras A, Moulonguet L, Billaud EM. Therapeutic drug monitoring in de novo kidney transplant receiving the modified-release once-daily tacrolimus. Transplant Proc. 2011;43:491-494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
6.  Hougardy JM, Broeders N, Kianda M, Massart A, Madhoun P, Le Moine A, Hoang AD, Mikhalski D, Wissing KM, Abramowicz D. Conversion from Prograf to Advagraf among kidney transplant recipients results in sustained decrease in tacrolimus exposure. Transplantation. 2011;91:566-569.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
7.  Kitada H, Okabe Y, Nishiki T, Miura Y, Kurihara K, Terasaka S, Kawanami S, Tuchimoto A, Masutani K, Tanaka M. One-year follow-up of treatment with once-daily tacrolimus in de novo renal transplant. Exp Clin Transplant. 2012;10:561-567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 10]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
8.  Busque S, Cantarovich M, Mulgaonkar S, Gaston R, Gaber AO, Mayo PR, Ling S, Huizinga RB, Meier-Kriesche HU. The PROMISE study: a phase 2b multicenter study of voclosporin (ISA247) versus tacrolimus in de novo kidney transplantation. Am J Transplant. 2011;11:2675-2684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 56]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
9.  Naidoo P, Rambiritch V. Voclosporin (ISA247) for plaque psoriasis. Lancet. 2008;372:888-889; author reply 889.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
10.  Papp K, Bissonnette R, Rosoph L, Wasel N, Lynde CW, Searles G, Shear NH, Huizinga RB, Maksymowych WP. Efficacy of ISA247 in plaque psoriasis: a randomised, multicentre, double-blind, placebo-controlled phase III study. Lancet. 2008;371:1337-1342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 54]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
11.  Bissonnette R, Papp K, Poulin Y, Lauzon G, Aspeslet L, Huizinga R, Mayo P, Foster RT, Yatscoff RW, Maksymowych WP. A randomized, multicenter, double-blind, placebo-controlled phase 2 trial of ISA247 in patients with chronic plaque psoriasis. J Am Acad Dermatol. 2006;54:472-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 37]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
12.  Kuglstatter A, Mueller F, Kusznir E, Gsell B, Stihle M, Thoma R, Benz J, Aspeslet L, Freitag D, Hennig M. Structural basis for the cyclophilin A binding affinity and immunosuppressive potency of E-ISA247 (voclosporin). Acta Crystallogr D Biol Crystallogr. 2011;67:119-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 33]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
13.  Gregory CR, Kyles AE, Bernsteen L, Wagner GS, Tarantal AF, Christe KL, Brignolo L, Spinner A, Griffey SM, Paniagua RT. Compared with cyclosporine, ISATX247 significantly prolongs renal-allograft survival in a nonhuman primate model. Transplantation. 2004;78:681-685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 21]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
14.  Aspeslet L, Freitag D, Huizinga R, Mayo P, Ling S, Foster R. The pharmacokinetics of voclosporin (VCS) in renal impairment. Am J Transpl. 2013;13 Suppl 5:abstr B1020.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Aspeslet L, Freitag D, Huizinga R, Mayo P, Ling S, Foster R. The effects of hepatic impairment on single and multiple oral dose pharmacokinetics of voclosporin (VCS). Am J Transpl. 2013;13 Suppl 5:abstr B1033.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Aspeslet L, Freitag D, Huizinga R, Mayo P, Ling S, Foster R. The effects of high fat breakfast, low fat breakfast or fasting on the absorption of voclosporin (VCS). Am J Transpl. 2013;13 Suppl 5:abstr B1032.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Alloway R, Bloom R, Gaston R, Goggins M, Mayo P, Busque S. A comparison of voclosporin and tacrolimus 1 year outcomes as predicted by PKPD in the PROMISE trial. Am J Transpl. 2013;13 Suppl 5:abstr B1018.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Vincenti F, Larsen C, Durrbach A, Wekerle T, Nashan B, Blancho G, Lang P, Grinyo J, Halloran PF, Solez K. Costimulation blockade with belatacept in renal transplantation. N Engl J Med. 2005;353:770-781.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 679]  [Cited by in F6Publishing: 602]  [Article Influence: 31.7]  [Reference Citation Analysis (0)]
19.  Vincenti F, Charpentier B, Vanrenterghem Y, Rostaing L, Bresnahan B, Darji P, Massari P, Mondragon-Ramirez GA, Agarwal M, Di Russo G. A phase III study of belatacept-based immunosuppression regimens versus cyclosporine in renal transplant recipients (BENEFIT study). Am J Transplant. 2010;10:535-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 722]  [Cited by in F6Publishing: 667]  [Article Influence: 47.6]  [Reference Citation Analysis (0)]
20.  Larsen CP, Grinyó J, Medina-Pestana J, Vanrenterghem Y, Vincenti F, Breshahan B, Campistol JM, Florman S, Rial Mdel C, Kamar N. Belatacept-based regimens versus a cyclosporine A-based regimen in kidney transplant recipients: 2-year results from the BENEFIT and BENEFIT-EXT studies. Transplantation. 2010;90:1528-1535.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 138]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
21.  Durrbach A, Pestana JM, Pearson T, Vincenti F, Garcia VD, Campistol J, Rial Mdel C, Florman S, Block A, Di Russo G. A phase III study of belatacept versus cyclosporine in kidney transplants from extended criteria donors (BENEFIT-EXT study). Am J Transplant. 2010;10:547-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 427]  [Cited by in F6Publishing: 387]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
22.  Florman S, Becker T, Bresnahan B, Chevaile-Ramos A, DeCarvalho D, Muehlbacher F, O’Connell P, Duan T, Agarwal M, Larsen C. Three year outcomes by donor type in phase III studies of belatacept vs cyclosporine in kidney transplantation (BENEFIT & BENEFIT-EXT). Am J Transplant. 2011;11 Suppl S2:abstract 229.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Ferguson R, Grinyó J, Vincenti F, Kaufman DB, Woodle ES, Marder BA, Citterio F, Marks WH, Agarwal M, Wu D. Immunosuppression with belatacept-based, corticosteroid-avoiding regimens in de novo kidney transplant recipients. Am J Transplant. 2011;11:66-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 139]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
24.  Rostaing L, Massari P, Garcia VD, Mancilla-Urrea E, Nainan G, del Carmen Rial M, Steinberg S, Vincenti F, Shi R, Di Russo G. Switching from calcineurin inhibitor-based regimens to a belatacept-based regimen in renal transplant recipients: a randomized phase II study. Clin J Am Soc Nephrol. 2011;6:430-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 135]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
25.  Vanrenterghem Y, Bresnahan B, Campistol J, Durrbach A, Grinyó J, Neumayer HH, Lang P, Larsen CP, Mancilla-Urrea E, Pestana JM. Belatacept-based regimens are associated with improved cardiovascular and metabolic risk factors compared with cyclosporine in kidney transplant recipients (BENEFIT and BENEFIT-EXT studies). Transplantation. 2011;91:976-983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 125]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
26.  Rostaing L, Neumayer HH, Reyes-Acevedo R, Bresnahan B, Florman S, Vitko S, Heifets M, Xing J, Thomas D, Vincenti F. Belatacept-versus cyclosporine-based immunosuppression in renal transplant recipients with pre-existing diabetes. Clin J Am Soc Nephrol. 2011;6:2696-2704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
27.  Oura T, Yamashita K, Suzuki T, Fukumori D, Watanabe M, Hirokata G, Wakayama K, Taniguchi M, Shimamura T, Miura T. Long-term hepatic allograft acceptance based on CD40 blockade by ASKP1240 in nonhuman primates. Am J Transplant. 2012;12:1740-1754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 53]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
28.  Goldwater R, Keirns J, Blahunka P, First R, Sawamoto T, Zhang W, Kowalski D, Kaibara A, Holman J. A phase 1, randomized ascending single-dose study of antagonist anti-human CD40 ASKP1240 in healthy subjects. Am J Transplant. 2013;13:1040-1046.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
29.  Vincenti F, Yang H, Klintmalm G, Steinberg S, Wang L, Zhang W, Conkle A, Blahunka P, First R, Holman J. Clinical Outcomes in a Phase 1b, Randomized, Double-Blind, Parallel Group, Placebo-Controlled, Single-Dose Study of ASKP1240 in De Novo Kidney Transplantation. Am J Transpl. 2013;13 Suppl 5:abstr 181.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Jordan SC, Vo AA, Tyan D, Nast CC, Toyoda M. Current approaches to treatment of antibody-mediated rejection. Pediatr Transplant. 2005;9:408-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 80]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
31.  Rocha PN, Butterly DW, Greenberg A, Reddan DN, Tuttle-Newhall J, Collins BH, Kuo PC, Reinsmoen N, Fields T, Howell DN. Beneficial effect of plasmapheresis and intravenous immunoglobulin on renal allograft survival of patients with acute humoral rejection. Transplantation. 2003;75:1490-1495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 134]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
32.  Becker YT, Becker BN, Pirsch JD, Sollinger HW. Rituximab as treatment for refractory kidney transplant rejection. Am J Transplant. 2004;4:996-1001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 263]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
33.  Montgomery R, Simpkins C, Zachary A. Anti-CD20 rescue therapy for kidneys undergoing antibody-mediated rejection. Am J Transplant. 2004;4:abstract 258.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Locke JE, Zachary AA, Haas M, Melancon JK, Warren DS, Simpkins CE, Segev DL, Montgomery RA. The utility of splenectomy as rescue treatment for severe acute antibody mediated rejection. Am J Transplant. 2007;7:842-846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 106]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
35.  Faguer S, Kamar N, Guilbeaud-Frugier C, Fort M, Modesto A, Mari A, Ribes D, Cointault O, Lavayssière L, Guitard J. Rituximab therapy for acute humoral rejection after kidney transplantation. Transplantation. 2007;83:1277-1280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 176]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
36.  Blume OR, Yost SE, Kaplan B. Antibody-mediated rejection: pathogenesis, prevention, treatment, and outcomes. J Transplant. 2012;2012:201754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
37.  Roberts DM, Jiang SH, Chadban SJ. The treatment of acute antibody-mediated rejection in kidney transplant recipients-a systematic review. Transplantation. 2012;94:775-783.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Lefaucheur C, Nochy D, Andrade J, Verine J, Gautreau C, Charron D, Hill GS, Glotz D, Suberbielle-Boissel C. Comparison of combination Plasmapheresis/IVIg/anti-CD20 versus high-dose IVIg in the treatment of antibody-mediated rejection. Am J Transplant. 2009;9:1099-1107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 203]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
39.  Schmidt N, Alloway RR, Walsh RC, Sadaka B, Shields AR, Girnita AL, Hanseman DJ, Woodle ES. Prospective evaluation of the toxicity profile of proteasome inhibitor-based therapy in renal transplant candidates and recipients. Transplantation. 2012;94:352-361.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
40.  Trivedi HL, Terasaki PI, Feroz A, Everly MJ, Vanikar AV, Shankar V, Trivedi VB, Kaneku H, Idica AK, Modi PR. Abrogation of anti-HLA antibodies via proteasome inhibition. Transplantation. 2009;87:1555-1561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 139]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
41.  Everly MJ, Terasaki PI, Hopfield J, Trivedi HL, Kaneku H. Protective immunity remains intact after antibody removal by means of proteasome inhibition. Transplantation. 2010;90:1493-1498.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
42.  Everly MJ, Everly JJ, Susskind B, Brailey P, Arend LJ, Alloway RR, Roy-Chaudhury P, Govil A, Mogilishetty G, Rike AH. Bortezomib provides effective therapy for antibody- and cell-mediated acute rejection. Transplantation. 2008;86:1754-1761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 335]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
43.  Flechner SM, Fatica R, Askar M, Stephany BR, Poggio E, Koo A, Banning S, Chiesa-Vottero A, Srinivas T. The role of proteasome inhibition with bortezomib in the treatment of antibody-mediated rejection after kidney-only or kidney-combined organ transplantation. Transplantation. 2010;90:1486-1492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 89]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
44.  Walsh RC, Everly JJ, Brailey P, Rike AH, Arend LJ, Mogilishetty G, Govil A, Roy-Chaudhury P, Alloway RR, Woodle ES. Proteasome inhibitor-based primary therapy for antibody-mediated renal allograft rejection. Transplantation. 2010;89:277-284.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 152]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
45.  Sberro-Soussan R, Zuber J, Suberbielle-Boissel C, Candon S, Martinez F, Snanoudj R, Rabant M, Pallet N, Nochy D, Anglicheau D. Bortezomib as the sole post-renal transplantation desensitization agent does not decrease donor-specific anti-HLA antibodies. Am J Transplant. 2010;10:681-686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 101]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
46.  Nigos JG, Arora S, Nath P, Hussain SM, Marcus RJ, Ko TY, Sureshkumar KK. Treatment of antibody-mediated rejection in kidney transplant recipients: a single-center experience with a bortezomib-based regimen. Exp Clin Transplant. 2012;10:609-613.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
47.  Tzvetanov I, Spaggiari M, Joseph J, Jeon H, Thielke J, Oberholzer J, Benedetti E. The use of bortezomib as a rescue treatment for acute antibody-mediated rejection: report of three cases and review of literature. Transplant Proc. 2012;44:2971-2975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
48.  Gheith O, Al-Otaibi T, Nampoory N, Halim M, Nair P, Saied T, Al-Waheeb S, Muzeirei I, Ibraheim M. Effective therapy for acute antibody-mediated rejection with mild chronic changes: case report and review of the literature. Exp Clin Transplant. 2012;10:406-409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
49.  Sureshkumar KK, Hussain SM, Marcus RJ, Ko TY, Khan AS, Tom K, Vivas CA, Parris G, Jasnosz KM, Thai NL. Proteasome inhibition with bortezomib: an effective therapy for severe antibody mediated rejection after renal transplantation. Clin Nephrol. 2012;77:246-253.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Hardinger KL, Murillo D. The influence of bortezomib on donor specific antibody reduction in patients with antibody mediated rejection. Clin Transpl. 2011;401-408.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Westphal S, Hansson S, Stelin G, Holgersson J, Mjörnstedt L, Friman S. Successful treatment of severe ABO antibody-mediated rejection using bortezomib: a case report. Transplant Proc. 2013;45:1213-1215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
52.  Waiser J, Budde K, Schütz M, Liefeldt L, Rudolph B, Schönemann C, Neumayer HH, Lachmann N. Comparison between bortezomib and rituximab in the treatment of antibody-mediated renal allograft rejection. Nephrol Dial Transplant. 2012;27:1246-1251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 92]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
53.  Rovira J, Cid J, Gutiérrez-García G, Pereira A, Fernández-Avilés F, Rosiñol L, Martínez C, Carreras E, Urbano A, Rovira M. Fatal immune hemolytic anemia following allogeneic stem cell transplantation: report of 2 cases and review of literature. Transfus Med Rev. 2013;27:166-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
54.  Roumenina LT, Loirat C, Dragon-Durey MA, Halbwachs-Mecarelli L, Sautes-Fridman C, Fremeaux-Bacchi V. Alternative complement pathway assessment in patients with atypical HUS. J Immunol Methods. 2011;365:8-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 113]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
55.  Châtelet V, Lobbedez T, Frémeaux-Bacchi V, Ficheux M, Ryckelynck JP, Hurault de Ligny B. Eculizumab: safety and efficacy after 17 months of treatment in a renal transplant patient with recurrent atypical hemolytic-uremic syndrome: case report. Transplant Proc. 2010;42:4353-4355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
56.  Al-Akash SI, Almond PS, Savell VH, Gharaybeh SI, Hogue C. Eculizumab induces long-term remission in recurrent post-transplant HUS associated with C3 gene mutation. Pediatr Nephrol. 2011;26:613-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 84]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
57.  Kavanagh D, Richards A, Goodship T, Jalanko H. Transplantation in atypical hemolytic uremic syndrome. Semin Thromb Hemost. 2010;36:653-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 38]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
58.  Zimmerhackl LB, Hofer J, Cortina G, Mark W, Würzner R, Jungraithmayr TC, Khursigara G, Kliche KO, Radauer W. Prophylactic eculizumab after renal transplantation in atypical hemolytic-uremic syndrome. N Engl J Med. 2010;362:1746-1748.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 127]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
59.  Peffault de Latour R, Xhaard A, Fremeaux-Bacchi V, Coppo P, Fischer AM, Helley D, Socié G. Successful use of eculizumab in a patient with post-transplant thrombotic microangiopathy. Br J Haematol. 2013;161:279-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 68]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
60.  Xie L, Nester CM, Reed AI, Zhang Y, Smith RJ, Thomas CP. Tailored eculizumab therapy in the management of complement factor H-mediated atypical hemolytic uremic syndrome in an adult kidney transplant recipient: a case report. Transplant Proc. 2012;44:3037-3040.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
61.  Zuber J, Le Quintrec M, Krid S, Bertoye C, Gueutin V, Lahoche A, Heyne N, Ardissino G, Chatelet V, Noël LH, Hourmant M, Niaudet P, Frémeaux-Bacchi V, Rondeau E, Legendre C, Loirat C; French Study Group for Atypical HUS. Eculizumab for atypical hemolytic uremic syndrome recurrence in renal transplantation. Am J Transplant. 2012;12:3337-3354.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Alachkar N, Bagnasco SM, Montgomery RA. Eculizumab for the treatment of two recurrences of atypical hemolytic uremic syndrome in a kidney allograft. Transpl Int. 2012;25:e93-e95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
63.  Krid S, Roumenina LT, Beury D, Charbit M, Boyer O, Frémeaux-Bacchi V, Niaudet P. Renal transplantation under prophylactic eculizumab in atypical hemolytic uremic syndrome with CFH/CFHR1 hybrid protein. Am J Transplant. 2012;12:1938-1944.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
64.  Wilson CH, Brown AL, White SA, Goodship TH, Sheerin NS, Manas DM. Successful treatment of de novo posttransplant thrombotic microangiopathy with eculizumab. Transplantation. 2011;92:e42-e43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
65.  Hadaya K, Ferrari-Lacraz S, Fumeaux D, Boehlen F, Toso C, Moll S, Martin PY, Villard J. Eculizumab in acute recurrence of thrombotic microangiopathy after renal transplantation. Am J Transplant. 2011;11:2523-2527.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 80]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
66.  Chandran S, Baxter-Lowe L, Olson JL, Tomlanovich SJ, Webber A. Eculizumab for the treatment of de novo thrombotic microangiopathy post simultaneous pancreas-kidney transplantation--a case report. Transplant Proc. 2011;43:2097-2101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
67.  Lonze BE, Dagher NN, Simpkins CE, Locke JE, Singer AL, Segev DL, Zachary AA, Montgomery RA. Eculizumab, bortezomib and kidney paired donation facilitate transplantation of a highly sensitized patient without vascular access. Am J Transplant. 2010;10:2154-2160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 57]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
68.  Cohney SJ, Hughes P, Rosemary M, Walker RG, Cantwell L, Vanhardeveld E, et al. C5 inhibition with eculizumab to prevent antibody mediated rejection (AbMR) in patients with donor specific anti-HLA antibody (DSAb) and a positive cross match. Am J Transplant. 2011;11 Suppl S2:abstr 1557.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Stegall MD, Diwan T, Raghavaiah S, Cornell LD, Burns J, Dean PG, Cosio FG, Gandhi MJ, Kremers W, Gloor JM. Terminal complement inhibition decreases antibody-mediated rejection in sensitized renal transplant recipients. Am J Transplant. 2011;11:2405-2413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 448]  [Cited by in F6Publishing: 421]  [Article Influence: 32.4]  [Reference Citation Analysis (0)]
70.  Biglarnia AR, Nilsson B, Nilsson T, von Zur-Mühlen B, Wagner M, Berne C, Wanders A, Magnusson A, Tufveson G. Prompt reversal of a severe complement activation by eculizumab in a patient undergoing intentional ABO-incompatible pancreas and kidney transplantation. Transpl Int. 2011;24:e61-e66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 63]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
71.  Stewart ZA, Collins TE, Schlueter AJ, Raife TI, Holanda DG, Nair R, Reed AI, Thomas CP. Case report: Eculizumab rescue of severe accelerated antibody-mediated rejection after ABO-incompatible kidney transplant. Transplant Proc. 2012;44:3033-3036.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
72.  Galliford J, Lawrence C, Willicombe M, Chan K, Roufosse C, McLean A, Cairns T, Cook HT. Reversal of refractory acute antibody mediated rejection with eculizumab. Am J Transplant. 2011;11 Suppl S2:abstr 1098.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  González-Roncero F, Suñer M, Bernal G, Cabello V, Toro M, Pereira P, Angel Gentil M. Eculizumab treatment of acute antibody-mediated rejection in renal transplantation: case reports. Transplant Proc. 2012;44:2690-2694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 42]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
74.  Kocak B, Arpali E, Demiralp E, Yelken B, Karatas C, Gorcin S, Gorgulu N, Uzunalan M, Turkmen A, Kalayoglu M. Eculizumab for salvage treatment of refractory antibody-mediated rejection in kidney transplant patients: case reports. Transplant Proc. 2013;45:1022-1025.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 36]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
75.  Locke JE, Magro CM, Singer AL, Segev DL, Haas M, Hillel AT, King KE, Kraus E, Lees LM, Melancon JK. The use of antibody to complement protein C5 for salvage treatment of severe antibody-mediated rejection. Am J Transplant. 2009;9:231-235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 267]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
76.  Locke JE; ClinicalTrials.  gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000- 2013; .  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Genentech , Inc . (2009-04-08). "Genentech Announces Voluntary Withdrawal of Raptiva from the U.S. Market". Press release.  Available from: http://www.drugs.com/news/genentech-announces-voluntary-raptiva-u-s-market-17125.html.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Vincenti F, Mendez R, Pescovitz M, Rajagopalan PR, Wilkinson AH, Butt K, Laskow D, Slakey DP, Lorber MI, Garg JP. A phase I/II randomized open-label multicenter trial of efalizumab, a humanized anti-CD11a, anti-LFA-1 in renal transplantation. Am J Transplant. 2007;7:1770-1777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 138]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
79.  Bashir SJ, Maibach HI. Alefacept (Biogen). Curr Opin Investig Drugs. 2001;2:631-634.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Weaver TA, Charafeddine AH, Agarwal A, Turner AP, Russell M, Leopardi FV, Kampen RL, Stempora L, Song M, Larsen CP. Alefacept promotes co-stimulation blockade based allograft survival in nonhuman primates. Nat Med. 2009;15:746-749.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 164]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
81.  Weaver TA; Astellas. (2009-12-15). "Voluntary US Market Discontinuation of Amevive (alefacept)". Press release.  Available from: http://www.amevive.com/. Retrieved June 6, 2013.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Rostaing L, Charpentier B, Glyda M, Rigotti P, Hettich F, Franks B, Houbiers JG, First R, Holman JM. Alefacept Combined With Tacrolimus, Mycophenolate Mofetil and Steroids in De Novo Kidney Transplantation: A Randomized Controlled Trial. Am J Transplant. 2013;13:1724-1733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
83.  Budde K, Sommerer C, Becker T, Asderakis A, Pietruck F, Grinyo JM, Rigotti P, Dantal J, Ng J, Barten MJ. Sotrastaurin, a novel small molecule inhibiting protein kinase C: first clinical results in renal-transplant recipients. Am J Transplant. 2010;10:571-581.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 67]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
84.  Transplantation: Development of sotrastaurin halted in kidney transplantation Nat Rev Nephrol. 2013;May 28; Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
85.  Friman S, Arns W, Nashan B, Vincenti F, Banas B, Budde K, Cibrik D, Chan L, Klempnauer J, Mulgaonkar S. Sotrastaurin, a novel small molecule inhibiting protein-kinase C: randomized phase II study in renal transplant recipients. Am J Transplant. 2011;11:1444-1455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 69]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
86.  Tedesco-Silva H, Kho MM, Hartmann A, Vitko S, Russ G, Rostaing L, Budde K, Campistol JM, Eris J, Krishnan I. Sotrastaurin in calcineurin inhibitor-free regimen using everolimus in de novo kidney transplant recipients. Am J Transplant. 2013;13:1757-1768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
87.  Russ GR, Tedesco-Silva H, Kuypers DR, Cohney S, Langer RM, Witzke O, Eris J, Sommerer C, von Zur-Mühlen B, Woodle ES. Efficacy of sotrastaurin plus tacrolimus after de novo kidney transplantation: randomized, phase II trial results. Am J Transplant. 2013;13:1746-1756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 35]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
88.  Busque S, Leventhal J, Brennan DC, Steinberg S, Klintmalm G, Shah T, Mulgaonkar S, Bromberg JS, Vincenti F, Hariharan S. Calcineurin-inhibitor-free immunosuppression based on the JAK inhibitor CP-690,550: a pilot study in de novo kidney allograft recipients. Am J Transplant. 2009;9:1936-1945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 121]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
89.  Vincenti F, Tedesco Silva H, Busque S, O’Connell P, Friedewald J, Cibrik D, Budde K, Yoshida A, Cohney S, Weimar W. Randomized phase 2b trial of tofacitinib (CP-690,550) in de novo kidney transplant patients: efficacy, renal function and safety at 1 year. Am J Transplant. 2012;12:2446-2456.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 125]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
90.  Busque S, Vincenti F, Tedesco Silva H, O’Connell P, Tortorici M, Lawendy N, Wang N, Chan G. Adverse events of over-immunosuppression are dependent on tofacitinib exposure in kidney transplant (KT) patients. Am J Transpl. 2013;13 Suppl 5:abstr 182.  [PubMed]  [DOI]  [Cited in This Article: ]