Editorial Open Access
Copyright ©2012 Baishideng. All rights reserved.
World J Transplant. Apr 24, 2012; 2(2): 19-26
Published online Apr 24, 2012. doi: 10.5500/wjt.v2.i2.19
Antibody induction therapy in adult kidney transplantation: A controversy continues
Kanwaljit K Chouhan, Rubin Zhang, Section of Nephrology, Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
Author contributions: Chouhan KK contributed to the design of the paper, acquisition, analysis and interpretation of the data from various studies which are reviewed in this paper, she also contributed to the final approval of the version to be published; Zhang R contributed by drafting the paper and revised it critically for important intellectual content, he also contributed to the final approval of the version to be published.
Correspondence to: Rubin Zhang, MD, FASN, Professor of Medicine, Medical Director of Kidney and Pancreas Transplantation, Tulane University Abdominal Transplant Institute, 1415 Tulane Ave, TW-35, New Orleans, LA 70112, United States. rzhang@tulane.edu
Telephone: +1-504-9881457 Fax: +1-504-9881105
Received: June 24, 2011
Revised: March 14, 2012
Accepted: March 20, 2012
Published online: April 24, 2012

Abstract

Antibody induction therapy is frequently used as an adjunct to the maintenance immunosuppression in adult kidney transplant recipients. Published data support antibody induction in patients with immunologic risk to reduce the incidence of acute rejection (AR) and graft loss from rejection. However, the choice of antibody remains controversial as the clinical studies were carried out on patients of different immunologic risk and in the context of varying maintenance regimens. Antibody selection should be guided by a comprehensive assessment of immunologic risk, patient comorbidities, financial burden as well as the maintenance immunosuppressives. Lymphocyte-depleting antibody (thymoglobulin, ATGAM or alemtuzumab) is usually recommended for those with high risk of rejection, although it increases the risk of infection and malignancy. For low risk patients, interleukin-2 receptor antibody (basiliximab or daclizumab) reduces the incidence of AR without much adverse effects, making its balance favorable in most patients. It should also be used in the high risk patients with other medical comorbidities that preclude usage of lymphocyte-depleting antibody safely. There are many patients with very low risk, who may be induced with intravenous steroids without any antibody, as long as combined potent immunosuppressives are kept as maintenance. In these patients, benefits with antibody induction may be too small to outweigh its adverse effects and financial cost. Rituximab can be used in desensitization protocols for ABO and/or HLA incompatible transplants. There are emerging data suggesting that alemtuzumab induction be more successful than other antibody for promoting less intensive maintenance protocols, such as steroid withdrawal, tacrolimus monotherapy or lower doses of tacrolimus and mycophenolic acid. However, the long-term efficacy and safety of these unconventional strategies remains unknown.

Key Words: Induction, Kidney transplant, Thymoglobulin, Basiliximab, Alemtuzumab, Acute rejection, Graft survival



INTRODUCTION

Appropriate immunosuppression is a key component of successful kidney transplantation. It is generally accepted that more intensive immunosuppression is required initially to prevent acute rejection (AR) and graft loss from AR, and less immunosuppression is subsequently maintained to allow the recipient to tolerate allograft and to minimize the adverse effects of immunosuppressive drugs. Many transplant centers in the USA routinely use an antilymphocyte antibody peri-operatively as induction therapy in addition to a maintenance regimen. In the year of 2008, 81.5% of kidney transplant recipients were given one of the following antibody inductions: thymoglobulin (44.8%), basiliximab (17.8%), daclizumab (10.9%), alemtuzumab (10.7%), and other 18.5% of patients do not receive any antibody induction[1]. The modern maintenance typically consists of a combination of two of the three classes of agents, calcineurin inhibitor (CNI, tacrolimus or cyclosporine), mycophenolic acid (mycophenolate mofetil or enteric coated mycophenolate sodium) and mammalian target of rapamycin inhibitor (sirolimus or everolimus), with or without steroids[1]. In this review, we will discuss the controversial issue of various antibody induction therapies, which were studied on adult patients of different immunologic risk in the context of varying maintenance immunosuppressive regimens.

OKT-3

OKT-3 is a murine monoclonal antibody against CD3 molecule. It depletes T cells by binding to the T-cell receptor-associated CD3 glycoprotein. Though historically used, it was never approved in the USA by the food and drug administration (FDA) as an induction agent. OKT-3 is associated with many side effects, including first-dose effect[2], pulmonary edema[3], nephropathy[4], infection[5,6] and malignancy[7]. Antithymocyte globulin (ATG) preparations were demonstrated to be superior than OKT-3 in terms of decrease in the incidence of AR and better tolerability[8-10]. The use of OKT-3 was subsequently decreased and led to cessation of its production in 2009.

ATG

There are two forms of ATG that are polyclonal immunoglobulins against human thymocytes from either horses (ATGAM) or rabbits (thymoglobulin). ATG binds to various cell surface markers, including CD2, CD3, CD4, CD8, CD11a and CD18, and leads to complement dependent lysis of lymphocytes. ATG as well as OKT-3 and alemtuzmab are often referred as lymphocyte-depleting antibodies. ATGAM was approved by FDA for both treatment and prevention of AR whereas thymoglobulin was only approved to treat AR episodes. ATG use is associated with cytokine release syndrome, myelosuppression and rarely anaphylactic reaction[11]. Several studies found that thymoglobulin was more effective in preventing AR and was associated with better graft survival than ATGAM[12-14]. Subsequently, ATGAM was used less frequently as induction therapy.

Dose of thymoglobulin induction has ranged from 1 to 4 mg/kg per day for 3 to 10 d. One study compared 3-d induction regimen (n = 40) with the historic 7-d course (n = 48). With 3-d course, thymoglobulin was administered at 3 mg/kg intra-operatively followed by 1.5 mg/kg on post-operative day 2 and 3. The 7-d course consisted of 1.5 mg/kg intra-operatively followed by same daily dose for next 6 d. Shorter initial hospital stay (6.1 d vs 8 d) and more profound lymphocyte depletion were observed in the 3-d group[15]. There was no difference in AR (5% vs 4.2%), graft survival (95% vs 98%) and patient survival (95% vs 98 %) at the end of 1 year in the 3-d vs 7-d group. Intraoperative administration of thymoglobulin was found to be associated with a lower incidence of delayed graft function (DGF) and shorter hospital stay[16]. Doses less than 3 mg/kg may not effectively prevent AR[16]. Higher dose and longer duration of induction was associated with increased risk of infection and lymphoma[17-21]. Therefore, the optimal dose of thymoglobulin induction might be a total of 6 mg/kg administered as 1.5 mg/kg per day in 3 to 5 d[17-21].

To compare thymoglobulin vs placebo induction, 89 sensitized renal transplant recipients received induction with (47 patients) or without (42 patients) thymoglobulin. The maintenance regimen consisted of cyclosporin, steroids and azathioprine. At the end of 1 year, the incidence of AR was 38% in thymoglobulin group and 64% in the placebo group. Both graft survival (89% vs 76%) and graft function were better in thymoglobulin group than the placebo group[22]. Similar benefits with ATG induction were reported by a meta-analysis of seven comparative studies[23]. Further analysis indicated that ATG induction might reduce the risk of graft loss greater in sensitized patients with high panel-reactive antibody (PRA) than in unsensitized patients[24].

These studies were performed in the era of less potent old maintenance immunosuppressives. The introduction of modern more potent maintenance drugs has successfully decreased the incidence of rejection and has improved graft survival[25-28]. The independent use of either mycophenolic acid[25,26] or tacrolimus[27,28] was found to have advantages over azathioprine or cyclosporine, respectively. In a 3-group comparative study with 6-mo follow up, AR was highest in the group receiving tacrolimus, azathioprine and prednisone without induction (25.4%) compared to the group receiving tacrolimus, azathioprine, prednisone and thymoglobulin induction (15.1%) and the group receiving cyclosporine, azathioprine, prednisone and thymoglobulin (21.2%)[29]. In the two thymoglobulin induction groups, tacrolimus arm had a lower incidence of AR than cyclosporine arm. The patient and graft survival were similar in all three groups. Both thymoglobulin groups had more side effects including leukopenia, thrombocytopenia and CMV infection. In the era of modern potent maintenance regimen including tacrolimus and mycophenolic acid, it is unlikely that ATG induction can still provide that much benefits as it was previously demonstrated in the context of less potent maintenance of cyclosporine and azathioprine.

INTERLEUKIN-2 RECEPTOR ANTIBODY

Daclizumab and basiliximab are the two interleukin (IL)-2 receptor antibodies (IL-2R Ab). Daclizumab is a humanized antibody and basiliximab is a chimeric monoclonal antibody. Both bind to the α chain of IL-2 receptor complex (CD25) expressed on activated T lymphocytes. This prevents the T cell activation and proliferation without causing cell lysis. Therefore, they are also known as non-depleting antibodies. IL-2R Ab was first introduced in 1997 and was FDA approved for induction therapy. They have the best safety profile compared to other available induction antibody without increased risk of infection or malignancy[30-32].

IL-2R Abs have been subjected to numerous placebo-controlled, randomized trials, which have showed a reduction in AR rate compared with placebo (28% vs 42%)[33-36]. In a meta analysis, the risk of AR is significantly reduced in patients who received IL-2R Ab induction than in those with placebo at 6 mo (12 trials: relative risk 0.66, 95% CI: 0.59-0.74) and at 1 year (10 trials: relative risk 0.67, 95% CI: 0.60-0.75)[37]. The incidences of CMV infection and malignancy at 1 year were similar to placebo control[37]. Both IL-2R Abs have similar efficacy and safety profile, but basiliximab is administered as 2 doses within 4 d of transplantation, whereas daclizumab is administered as 5 doses over 8 wk[19,32]. This difference in convenience of administration led to more frequent use of basiliximab than daclizumab. Subsequently, Roche pharmaceuticals withdrew daclizumab from market in October 2008.

Our center’s decade - long experience has indicated that basiliximab induction is safe and adequate for kidney transplant, including the high risk transplants, such as deceased donor kidney transplants in highly sensitized African Americans[38], simultaneous kidney pancreas transplant in African Americans[39] and splitting single pediatric donor kidney transplant[40], as long as the conventional triple regimen consisting of tacrolimus, mycophenolic acid and steroids are used as maintenance. A recent analysis based on USRDS data from 2000 to 2005 also indicated that both patient and graft survival were similar in African Americans and Caucasian patients using either thymoglobulin or IL-2R Ab induction[41].

IL-2 RECEPTOR ANTIBODY VS ATG

The safety and efficacy of thymoglobulin and basiliximab induction were compared in 278 high risk patients who received deceased donor kidneys[42]. High risk was determined according to the duration of cold ischemia and various other donor and recipient risk factors including donor age > 50 years, donation after cardiac death, donor with ATN or requiring high dose of ionotropic support, repeat transplant, PRA > 20%, black race and one or more HLA mismatches. Both groups received cyclosporine, mycophenolate mofetil and prednisone as maintenance. At 12 mo, there were fewer biopsy-proven AR in the thymoglobulin group than in the basiliximab group (15.6% vs 25.5%, P = 0.02). Severe rejection, as indicated by the need for antibody treatment, was less frequent in thymoglobulin group than basiliximab group (1.4% vs 8.0%, P = 0.005). The incidence of DGF (40.4% vs 44.5%, P = 0.54), graft loss (9.2% vs 10.2%) and death (4.3% vs 4.4%) was similar in both groups. However the incidences of infection and malignancy were significantly higher in thymoglobulin group than basiliximab group. A 5-year follow-up of these patients showed that AR remained lower in thymoglobulin than basiliximab group, but graft and patient survival were still not different[43]. Similar result was also reported by Noël et al[44] in 227 high risk patients who received modern maintenance of tacrolimus, mycophenolate mofetil and corticosteroid. High risk was defined as PRA > 30% and/or peak PRA > 50%, loss of a first renal transplant to rejection within 2 years or history of two or more previous transplants. Compared to the daclizumab group, thymoglobulin group had lower incidence of AR (15.0% vs 27.2%), and steroid-resistant AR (2.7% vs 14.9%) and also delayed the time to AR (35 d vs 13 d) in 1 year[44]. However, there was no difference in either graft or patient survival. The number of bacterial infection per patient (2.5 ± 1.8 vs 1.7 ± 1.2, P = 0.01) and the incidence of CMV infection (18.6% vs 10.5%, P = 0.09) was significantly higher in the thymoglobulin group than in the daclizumab group. These clinical trials show that thymoglobulin induction reduces the risk of AR, but it increases the risk of infection and possible malignancy. There is no convincing clinical evidence of superior graft or patient survival with thymoglobulin induction than the IL-2R antibody induction in high-risk patients.

Using SRTR database, Patlolla et al[45] analyzed a total of 48 948 recipients of first renal transplants who were discharged on CNI (cyclosporine or tacrolimus) and anti-metabolite (mycophenolic acid or azathioprine). Induction with IL-2R Ab (basiliximab or daclizumab, n = 17 472) was associated with a reduction in both AR (odds ratio 0.81, 95% CI: 0.75-0.87) and graft loss (hazard ratio 0.90, 95% CI: 0.84-0.95) compared with no antibody induction (n = 22 008). The greater the HLA mismatch, higher the efficacy of IL-2R Ab in reducing AR. Compared to IL-2R Ab induction, lymphocyte - depleting antibody (thymoglobulin, ATGAM or OKT-3, n = 9468) was associated with lower risk of AR (OR: 0.90, 95% CI: 0.83-0.99) at 1 year, but not associated with any better graft survival (OR 1.08, 95% CI: 1.00-1.18). Several studies directly compared thymoglobulin with IL-2R Ab induction in patients with low immunologic risk[46-48]. Similar rejection rate and graft survival, but higher incidence of infection was reported in those received thymoglobulin than IL-2R Ab induction. These clinical data, taken together with other trials comparing IL-2R Ab induction with placebo[33-37] supports use of IL-2R Ab rather than thymoglobulin for induction in low risk patients.

ALEMTUZUMAB

Alemtuzumab is a humanized anti-CD52 monoclonal antibody, which triggers the antibody-dependent lysis of lymphocytes (both B and T cells), monocytes and NK cells. Alemtuzumab is FDA approved for treating B cell lymphomas. It was first introduced to kidney transplant by Calne et al[49] in late 1990s. As an induction agent, it produces a profound depletion of lymphocytes and is associated with more frequent and severe adverse effects, such as neutropenia, thrombocytopenia, autoimmune hemolytic anemia and other autoimmune diseases[50,51]. However, it did not appear to affect the incidence of recurrent glomerulonephritis[52]. Two doses of alemtuzumab were initially administrated for induction[53,54]. Due to its profound immunosuppression, single dose (30 mg, given intraoperatively) has been subsequently studied[55,56]. It is also hoped that alemtuzumab induction could permit patients to be maintained on less intensive immunosuppression, such as tacrolimus monotherapy[57,58], steroid-free regimen[59,60], or lower doses of tacrolimus and mycophenolic acid[59,61].

Margreiter et al[57] assessed the efficacy of alemtuzumab induction with tacrolimus monotherapy (n = 65) as compared to no induction with tacrolimus, mycophenolate mofetil and steroid maintenance (n = 66) for deceased donor kidney transplant. At 12 mo, the incidence of AR was not statistically different (20% vs 32%, P = 0.09). The graft and patient survival were similar, but alemtuzumab group had more CMV infection. This protocol was also studied in living donor kidney transplant by Tan et al[62,63]. A total of 205 living donor recipients were treated with alemtuzumab induction followed by tacrolimus monotherapy and 47 controls were treated with conventional triple therapy of mycophenolate, tacrolimus and prednisone without induction. At 1 year, the incidence of AR was much lower in the alemtuzumab group (6.8% vs 17%, P < 0.05)[62]. The 1, 2, and 3-year patient survival (99%, 98% and 96.4%) and the graft survival (90.8%, 93.3% and 86.3%) in the alemtuzumab group, are similar to the SRTR data for living donor kidney transplantation[63].

Induction with alemtuzumab (n = 123) and basiliximab (n = 155) were compared in a steroid-free maintenance consisting of mycophenolate acid and tacrolimus[64]. Early rejection (< 3 mo) rates were higher in the basiliximab group (11.6% vs 4.1%) but were equal at 1 year in the two groups (13.5% vs 14.9%, P = NS). The 1-year death censored graft survival was 99.2% for the alemtuzumab and 99.4% in the basiliximab group (P = NS). The incidence of CMV disease (4% vs 5%) and malignancy (2 recipients in each group) were also similar in the two groups. Therefore, in steroid-free maintenance, alemtuzumab induction is associated with lower incidence of early rejection, but similar graft survival compared to basiliximab induction.

ALEMTUZUMAB VS BASILIXIMAB VS THYMOGLOBULIN

These three antibody induction agents were first compared by Ciancio et al[59] in 90 deceased donor kidney transplants. Maintenance immunosuppression was tacrolimus (target trough level of 8-10 ng/mL), mycophenolate mofetil (1000 mg twice daily) and prednisone in thymoglobulin and daclizumab groups, while alemtuzumab group received lower doses of tacrolimus (target trough level of 4-7 ng/mL) and mycophenolate mofetil (500 mg twice daily). At 1 year, there was no significant difference in the three groups for AR, graft survival or patient survival. At 2 years, cumulative incidences of AR were 20%, 23% and 23% in thymoglobulin, alemtuzumab and daclizumab groups, respectively[61]. The overall patient and graft survival were similar, but there was a trend towards worse death censored graft survival and more chronic allograft nephropathy in alemtuzumab group[61]. In another study of rapid steroid withdrawal in a total of 474 kidney recipients, 139 high risk patients (African American, PRA ≥ 20% or re-transplants) were induced with alemtuzumab or thymoglobulin, while 335 low risk patients (non African American, PRA < 20% or primary transplant) were induced with alemtuzumab or basiliximab[60]. At 2 years, alemtuzumab induction has lower incidence of AR than basiliximab (8.9% vs 21.7%, P < 0.05) for low risk patients. The high-risk patients experienced same rejection rates with either thymoglobulin or alemtuzumab induction (13% in both groups). Patient and graft survival at 2 years were similar between the groups in both high risk patients (98.6% vs 93% and 92.2% vs 88.4%, alemtuzumab vs thymoglobulin, P = NS) and low risk patients (97.4% vs 98% and 96.2% vs 92.3%, alemtuzumab vs basiliximab, P = NS). A 3-year follow up showed similar results in terms of lower incidence of AR with alemtuzumab than basiliximab (10% vs 22%, P = 0.003) in low risk patients, while no difference in AR between alemtuzumab and thymoglobulin (18% vs 15%, P = 0.63) in high risk patients[65].

From these data, alemtuzumab induction appears to be more successful than other induction for unconventional protocols, such as steroid withdrawal, tacrolimus monotherapy or lower doses of tacrolimus and mycophenolic acid. However, these studies are small, short-term and should be considered as experimental. The long-term efficacy of these protocols remains to be vigorously investigated[53,61,66]. One obvious concern is that lymphocytes could recover from the initial depletion if insufficient maintenance immunosuppression is left over long term, which potentially leads to development of AR and/or chronic rejection. Late development of de novo donor specific antibodies (DSA) is increasingly recognized as an independent and detrimental factor for accelerated transplant glomerulopathy and graft loss[67,68].

RITUXIMAB

Rituximab is a chimeric monoclonal Ab against CD20, which is expressed on the majority of B cells. It was first approved in 1997 for refractory B cell lymphomas and it is increasingly applied for autoimmune diseases. In the realm of kidney transplant, rituximab has been used for the treatment of AMR and desensitization in ABO and/or HLA incompatible transplants[69,70]. Takagi et al[70] reported rituximab induction in desensitization of 78 ABO and/or HLA incompatible transplants, all of them also received 3-4 sessions of double-filtration plasmapheresis before transplant. Compared with the non-rituximab group of 66 compatible transplants, rituximab group had significantly lower incidence of ACR (8.2% vs 23.3%, P < 0.05), but not higher incidence of AMR (6.8% vs 8.3%, P = 0.75). Anti-HLA Ab to class 1 and class 2 were depleted by 70% and 83%, respectively for more than 2 years after rituximab induction. The incidences of CMV infection (26% vs 29%, P = 1.0) or leukopenia (23% vs 14%, P = 0.25) were not different, and the 2-year survival rates of patient (100% vs 98%, P = 0.28) and graft (99% vs 100%, P = 0.91) were excellent in both groups[70]. Therefore, rituximab appears to be a safe and effective induction Ab for the desensitization protocol of ABO or HLA incompatible transplants.

In the setting of non-desensitization, Clatworthy et al[71] reported that 5 of 6 patients (83%) induced with rituximab had ACR in the first 3 mo after transplant as compared with 1 of 7 patients (14%) induced with daclizumab (P = 0.01). However, Tydén et al[72] reported a randomized, doubleblind multicenter study that included 68 rituximab and 68 placebo patients. All patients received conventional maintenance of tacrolimus, mycophenolic acid and steroids. During the first 6 mo, there were 10 treatment failure (defined as AR, graft loss or death) in rituximab group vs 14 in placebo group (P = 0.35). There was a tendency toward fewer AR (8/68 vs 12/68, P = 0.32) and milder AR without increase in infections or leukopenia in the rituximab group. Long-term study is needed to further determine the benefits of rituximab induction for non-sensitized patients.

OTHER CONSIDERATION

Apart from the immunologic risk, many other medical and physical factors should also be considered in the choice of induction therapy. Depleting antibody (thymoglobulin, ATGAM, OKT-3 and alemtuzumab) induction should be avoided in patients with history of malignancy, severe viral infection (including HIV, HBV or HCV), hematological disorder of leucopenia or thrombocytopenia and elderly with cardiac or pulmonary comorbidities[73-75]. For these patients, we do not use any antibody induction if they do not have high immunologic risk, and we use IL-2R Ab induction (not lymphocyte depleting antibody) for those who do have high immunologic risk. A recent study of 150 HIV-infected patients who underwent kidney transplant indicated that ATG induction significantly increased the risk of graft loss (HR 2.5, 95% CI: 1.1-5.6, P = 0.03). ATG induction was also associated with twice as many serious infections per follow-up year as patients received IL2R Ab induction or no induction (0.9 vs 0.4, P = 0.002)[74]. Another study reported that 2-year patient survival was less than 50% in the elderly (more than 60 years old) who had DGF and received thymoglobulin induction[75].

The financial costs of antibody induction therapies are significantly different. In US, the average whole sale price for the typical dose of alemtuzumab (30 mg × 1 dose) is $1982.70; basiliximab (20 mg × 2 doses) is $5338.66; while thymoglubulin (1.5 mg/kg × 4 doses for a 70 kg patient) costs $10 200.00[76]. A financial analysis indicated that IL-2R Ab (basiliximab/daclizumab) was more cost effective than placebo (no induction) or induction with lymphocyte-depleting antibody (OKT3/ATG/ATGAM)[77].

CONCLUSION

Published data support the usage of antibody induction therapy in adult patient with immunologic risk to reduce the incidence of AR and possible graft loss from rejection. However, the choice of antibody remains controversial. Antibody selection should be guided by a comprehensive assessment of immunologic risk of recipient and donor organ, patient comorbidities, financial burden, and more importantly, the maintenance immunosuppressive regimen. Lymphocyte-depleting antibody is recommended for those with high immunologic risk as outlined in the 2009 KDIGO clinical practice guidelines[78] (sensitized patient, presence of DSA, ABO incompatibility, high HLA mismatches, DGF, cold ischemia time > 24 h, African-American ethnicity, younger recipient age, older donor age), though it increases the risk of infection and malignancy. For low risk patients, IL-2R Ab induction reduces the incidence of AR and graft loss without much adverse effects, making its balance favorable in most patients. IL-2R Ab induction should also be used in the high risk patients with other medical comorbidities that preclude usage of any lymphocyte-depleting antibody safely. We believe that many patients with very low risk (non-sensitized, Caucasian, Asian, well HLA matched, living related donor transplant) may be induced with intravenous steroids without using any antibody, as long as combined potent immunosuppressives are kept as maintenance. In these patients, benefits with antibody induction may be too small to outweigh its adverse effects and the financial cost. Rituximab induction is useful in desensitization protocols for ABO and/or HLA incompatible transplants. Alemtuzumab induction might be more successful than other antibody induction for adopting less intensive maintenance protocols, such as steroids withdrawal, tacrolimus monotherapy or lower doses of tacrolimus and mycophenolic acid. However, the long-term safety and efficacy of these unconventional strategies remains to be determined.

Footnotes

Peer reviewers: Costas Fourtounas, MD, PhD, Associate Professor, Department of Nephrology, Patras University Hospital, Rio-Patras 26500, Greece; Caigan Du, PhD, Assistant Professor, Department of Urologic Sciences, University of British Columbia, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada

S- Editor Cheng JX L- Editor A E- Editor Zheng XM

References
1.  US Department of Health and Human Services. OPTN/SRTR Annual Report. 2009.  Available from: http://www.ustransplant.org/annual_reports/current/506a_ki.htm.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Kirk AD. Induction immunosuppression. Transplantation. 2006;82:593-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 93]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
3.  Hardinger KL, Koch MJ, Brennan DC. Current and future immunosuppressive strategies in renal transplantation. Pharmacotherapy. 2004;24:1159-1176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
4.  First MR, Schroeder TJ, Hariharan S. OKT3-induced cytokine-release syndrome: renal effects (cytokine nephropathy). Transplant Proc. 1993;25:25-26.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Hibberd PL, Tolkoff-Rubin NE, Cosimi AB, Schooley RT, Isaacson D, Doran M, Delvecchio A, Delmonico FL, Auchincloss H, Rubin RH. Symptomatic cytomegalovirus disease in the cytomegalovirus antibody seropositive renal transplant recipient treated with OKT3. Transplantation. 1992;53:68-72.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Thistlethwaite JR, Stuart JK, Mayes JT, Gaber AO, Woodle S, Buckingham MR, Stuart FP. Complications and monitoring of OKT3 therapy. Am J Kidney Dis. 1988;11:112-119.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Swinnen LJ, Costanzo-Nordin MR, Fisher SG, O'Sullivan EJ, Johnson MR, Heroux AL, Dizikes GJ, Pifarre R, Fisher RI. Increased incidence of lymphoproliferative disorder after immunosuppression with the monoclonal antibody OKT3 in cardiac-transplant recipients. N Engl J Med. 1990;323:1723-1728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 715]  [Cited by in F6Publishing: 722]  [Article Influence: 21.2]  [Reference Citation Analysis (0)]
8.  Jaffers GJ, Fuller TC, Cosimi AB, Russell PS, Winn HJ, Colvin RB. Monoclonal antibody therapy. Anti-idiotypic and non-anti-idiotypic antibodies to OKT3 arising despite intense immunosuppression. Transplantation. 1986;41:572-578.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Chatenoud L, Baudrihaye MF, Chkoff N, Kreis H, Goldstein G, Bach JF. Restriction of the human in vivo immune response against the mouse monoclonal antibody OKT3. J Immunol. 1986;137:830-838.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Kumar MS, Heifets M, Moritz MJ, Saeed MI, Khan SM, Fyfe B, Sustento-Riodeca N, Daniel JN, Kumar A. Safety and efficacy of steroid withdrawal two days after kidney transplantation: analysis of results at three years. Transplantation. 2006;81:832-839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 76]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
11.  Micromedex healthcare series. Greenwood Village, CO: Thomson Micromedex Inc. 2009; .  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Brennan DC, Flavin K, Lowell JA, Howard TK, Shenoy S, Burgess S, Dolan S, Kano JM, Mahon M, Schnitzler MA. A randomized, double-blinded comparison of Thymoglobulin versus Atgam for induction immunosuppressive therapy in adult renal transplant recipients. Transplantation. 1999;67:1011-1018.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Hardinger KL, Schnitzler MA, Miller B, Lowell JA, Shenoy S, Koch MJ, Enkvetchakul D, Ceriotti C, Brennan DC. Five-year follow up of thymoglobulin versus ATGAM induction in adult renal transplantation. Transplantation. 2004;78:136-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 103]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
14.  Hardinger KL, Rhee S, Buchanan P, Koch M, Miller B, Enkvetchakul D, Schuessler R, Schnitzler MA, Brennan DC. A prospective, randomized, double-blinded comparison of thymoglobulin versus Atgam for induction immunosuppressive therapy: 10-year results. Transplantation. 2008;86:947-952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 65]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
15.  Agha IA, Rueda J, Alvarez A, Singer GG, Miller BW, Flavin K, Lowell JA, Shenoy S, Howard TK, Ramachandran V. Short course induction immunosuppression with thymoglobulin for renal transplant recipients. Transplantation. 2002;73:473-475.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Goggins WC, Pascual MA, Powelson JA, Magee C, Tolkoff-Rubin N, Farrell ML, Ko DS, Williams WW, Chandraker A, Delmonico FL. A prospective, randomized, clinical trial of intraoperative versus postoperative Thymoglobulin in adult cadaveric renal transplant recipients. Transplantation. 2003;76:798-802.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Wong W, Agrawal N, Pascual M, Anderson DC, Hirsch HH, Fujimoto K, Cardarelli F, Winkelmayer WC, Cosimi AB, Tolkoff-Rubin N. Comparison of two dosages of thymoglobulin used as a short-course for induction in kidney transplantation. Transpl Int. 2006;19:629-635.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 51]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
18.  Stevens RB, Mercer DF, Grant WJ, Freifeld AG, Lane JT, Groggel GC, Rigley TH, Nielsen KJ, Henning ME, Skorupa JY. Randomized trial of single-dose versus divided-dose rabbit anti-thymocyte globulin induction in renal transplantation: an interim report. Transplantation. 2008;85:1391-1399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 43]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
19.  Micromedex healthcare series; Greenwood village, CO: Thompson healthcare inclusive, 2007.  .  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Beiras-Fernandez A, Thein E, Hammer C. Induction of immunosuppression with polyclonal antithymocyte globulins: an overview. Exp Clin Transplant. 2003;1:79-84.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Hardinger KL. Rabbit antithymocyte globulin induction therapy in adult renal transplantation. Pharmacotherapy. 2006;26:1771-1783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 74]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
22.  Thibaudin D, Alamartine E, de Filippis JP, Diab N, Laurent B, Berthoux F. Advantage of antithymocyte globulin induction in sensitized kidney recipients: a randomized prospective study comparing induction with and without antithymocyte globulin. Nephrol Dial Transplant. 1998;13:711-715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 70]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
23.  Szczech LA, Berlin JA, Aradhye S, Grossman RA, Feldman HI. Effect of anti-lymphocyte induction therapy on renal allograft survival: a meta-analysis. J Am Soc Nephrol. 1997;8:1771-1777.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Szczech LA, Berlin JA, Feldman HI. The effect of antilymphocyte induction therapy on renal allograft survival. A meta-analysis of individual patient-level data. Anti-Lymphocyte Antibody Induction Therapy Study Group. Ann Intern Med. 1998;128:817-826.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Sollinger HW. Mycophenolate mofetil for the prevention of acute rejection in primary cadaveric renal allograft recipients. U.S. Renal Transplant Mycophenolate Mofetil Study Group. Transplantation. 1995;60:225-232.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  A blinded, randomized clinical trial of mycophenolate mofetil for the prevention of acute rejection in cadaveric renal transplantation. The Tricontinental Mycophenolate Mofetil Renal Transplantation Study Group. Transplantation. 1996;61:1029-1037.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Knoll GA, Bell RC. Tacrolimus versus cyclosporin for immunosuppression in renal transplantation: meta-analysis of randomised trials. BMJ. 1999;318:1104-1107.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Krämer BK, Del Castillo D, Margreiter R, Sperschneider H, Olbricht CJ, Ortuño J, Sester U, Kunzendorf U, Dietl KH, Bonomini V. Efficacy and safety of tacrolimus compared with ciclosporin A in renal transplantation: three-year observational results. Nephrol Dial Transplant. 2008;23:2386-2392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 43]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
29.  Charpentier B, Rostaing L, Berthoux F, Lang P, Civati G, Touraine JL, Squifflet JP, Vialtel P, Abramowicz D, Mourad G. A three-arm study comparing immediate tacrolimus therapy with antithymocyte globulin induction therapy followed by tacrolimus or cyclosporine A in adult renal transplant recipients. Transplantation. 2003;75:844-851.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Mottershead M, Neuberger J. Daclizumab. Expert Opin Biol Ther. 2007;7:1583-1596.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
31.  Penfornis A, Kury-Paulin S. Immunosuppressive drug-induced diabetes. Diabetes Metab. 2006;32:539-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 90]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
32.  Ramirez CB, Marino IR. The role of basiliximab induction therapy in organ transplantation. Expert Opin Biol Ther. 2007;7:137-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 55]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
33.  Nashan B, Moore R, Amlot P, Schmidt AG, Abeywickrama K, Soulillou JP. Randomised trial of basiliximab versus placebo for control of acute cellular rejection in renal allograft recipients. CHIB 201 International Study Group. Lancet. 1997;350:1193-1198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 595]  [Cited by in F6Publishing: 613]  [Article Influence: 22.7]  [Reference Citation Analysis (0)]
34.  Vincenti F, Kirkman R, Light S, Bumgardner G, Pescovitz M, Halloran P, Neylan J, Wilkinson A, Ekberg H, Gaston R. Interleukin-2-receptor blockade with daclizumab to prevent acute rejection in renal transplantation. Daclizumab Triple Therapy Study Group. N Engl J Med. 1998;338:161-165.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Kahan BD, Rajagopalan PR, Hall M. Reduction of the occurrence of acute cellular rejection among renal allograft recipients treated with basiliximab, a chimeric anti-interleukin-2-receptor monoclonal antibody. United States Simulect Renal Study Group. Transplantation. 1999;67:276-284.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Lawen JG, Davies EA, Mourad G, Oppenheimer F, Molina MG, Rostaing L, Wilkinson AH, Mulloy LL, Bourbigot BJ, Prestele H. Randomized double-blind study of immunoprophylaxis with basiliximab, a chimeric anti-interleukin-2 receptor monoclonal antibody, in combination with mycophenolate mofetil-containing triple therapy in renal transplantation. Transplantation. 2003;75:37-43.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Webster AC, Playford EG, Higgins G, Chapman JR, Craig JC. Interleukin 2 receptor antagonists for renal transplant recipients: a meta-analysis of randomized trials. Transplantation. 2004;77:166-176.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Ren Q, Paramesh A, Yau CL, Killackey M, Slakey D, Florman S, Buell J, Alper B, Simon E, Hamm LL. Long-term outcome of highly sensitized African American patients transplanted with deceased donor kidneys. Transpl Int. 2011;24:259-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
39.  Zhang R, Florman S, Devidoss S, Zarifian A, Yau CL, Paramesh A, Killackey M, Alper B, Fonseca V, Slakey D. A comparison of long-term survivals of simultaneous pancreas-kidney transplant between African American and Caucasian recipients with basiliximab induction therapy. Am J Transplant. 2007;7:1815-1821.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
40.  Zhang R, Paramesh A, Florman S, Yau CL, Balamuthusamy S, Krane NK, Slakey D. Long-term outcome of adults who undergo transplantation with single pediatric kidneys: how young is too young. Clin J Am Soc Nephrol. 2009;4:1500-1506.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Jindal RM, Das NP, Neff RT, Hurst FP, Falta EM, Elster EA, Abbott KC. Outcomes in African-Americans vs. Caucasians using thymoglobulin or interleukin-2 receptor inhibitor induction: analysis of USRDS database. Am J Nephrol. 2009;29:501-508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
42.  Brennan DC, Daller JA, Lake KD, Cibrik D, Del Castillo D. Rabbit antithymocyte globulin versus basiliximab in renal transplantation. N Engl J Med. 2006;355:1967-1977.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 573]  [Cited by in F6Publishing: 526]  [Article Influence: 29.2]  [Reference Citation Analysis (0)]
43.  Brennan DC, Schnitzler MA. Long-term results of rabbit antithymocyte globulin and basiliximab induction. N Engl J Med. 2008;359:1736-1738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 92]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
44.  Noël C, Abramowicz D, Durand D, Mourad G, Lang P, Kessler M, Charpentier B, Touchard G, Berthoux F, Merville P. Daclizumab versus antithymocyte globulin in high-immunological-risk renal transplant recipients. J Am Soc Nephrol. 2009;20:1385-1392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 149]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
45.  Patlolla V, Zhong X, Reed GW, Mandelbrot DA. Efficacy of anti-IL-2 receptor antibodies compared to no induction and to antilymphocyte antibodies in renal transplantation. Am J Transplant. 2007;7:1832-1842.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Lebranchu Y, Bridoux F, Büchler M, Le Meur Y, Etienne I, Toupance O, Hurault de Ligny B, Touchard G, Moulin B, Le Pogamp P. Immunoprophylaxis with basiliximab compared with antithymocyte globulin in renal transplant patients receiving MMF-containing triple therapy. Am J Transplant. 2002;2:48-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 181]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
47.  Mourad G, Rostaing L, Legendre C, Garrigue V, Thervet E, Durand D. Sequential protocols using basiliximab versus antithymocyte globulins in renal-transplant patients receiving mycophenolate mofetil and steroids. Transplantation. 2004;78:584-590.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Kyllönen LE, Eklund BH, Pesonen EJ, Salmela KT. Single bolus antithymocyte globulin versus basiliximab induction in kidney transplantation with cyclosporine triple immunosuppression: efficacy and safety. Transplantation. 2007;84:75-82.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Calne R, Friend P, Moffatt S, Bradley A, Hale G, Firth J, Bradley J, Smith K, Waldmann H. Prope tolerance, perioperative campath 1H, and low-dose cyclosporin monotherapy in renal allograft recipients. Lancet. 1998;351:1701-1702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 358]  [Cited by in F6Publishing: 356]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
50.  Haider I, Cahill M. Fatal thrombocytopaenia temporally related to the administration of alemtuzumab (MabCampath) for refractory CLL despite early discontinuation of therapy. Hematology. 2004;9:409-411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 20]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
51.  Kirk AD, Hale DA, Swanson SJ, Mannon RB. Autoimmune thyroid disease after renal transplantation using depletional induction with alemtuzumab. Am J Transplant. 2006;6:1084-1085.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 55]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
52.  Pascual J, Mezrich JD, Djamali A, Leverson G, Chin LT, Torrealba J, Bloom D, Voss B, Becker BN, Knechtle SJ. Alemtuzumab induction and recurrence of glomerular disease after kidney transplantation. Transplantation. 2007;83:1429-1434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 27]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
53.  Watson CJ, Bradley JA, Friend PJ, Firth J, Taylor CJ, Bradley JR, Smith KG, Thiru S, Jamieson NV, Hale G. Alemtuzumab (CAMPATH 1H) induction therapy in cadaveric kidney transplantation--efficacy and safety at five years. Am J Transplant. 2005;5:1347-1353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 187]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
54.  Knechtle SJ, Pirsch JD, H Fechner J, Becker BN, Friedl A, Colvin RB, Lebeck LK, Chin LT, Becker YT, Odorico JS. Campath-1H induction plus rapamycin monotherapy for renal transplantation: results of a pilot study. Am J Transplant. 2003;3:722-730.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 324]  [Cited by in F6Publishing: 326]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
55.  Morris PJ, Russell NK. Alemtuzumab (Campath-1H): a systematic review in organ transplantation. Transplantation. 2006;81:1361-1367.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Ciancio G, Burke GW. Alemtuzumab (Campath-1H) in kidney transplantation. Am J Transplant. 2008;8:15-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 62]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
57.  Margreiter R, Klempnauer J, Neuhaus P, Muehlbacher F, Boesmueller C, Calne RY. Alemtuzumab (Campath-1H) and tacrolimus monotherapy after renal transplantation: results of a prospective randomized trial. Am J Transplant. 2008;8:1480-1485.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Thomas PG, Woodside KJ, Lappin JA, Vaidya S, Rajaraman S, Gugliuzza KK. Alemtuzumab (Campath 1H) induction with tacrolimus monotherapy is safe for high immunological risk renal transplantation. Transplantation. 2007;83:1509-1512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 60]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
59.  Ciancio G, Burke GW, Gaynor JJ, Carreno MR, Cirocco RE, Mathew JM, Mattiazzi A, Cordovilla T, Roth D, Kupin W. A randomized trial of three renal transplant induction antibodies: early comparison of tacrolimus, mycophenolate mofetil, and steroid dosing, and newer immune-monitoring. Transplantation. 2005;80:457-465.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Mulgaonkar S, Hanaway M, Woodle ES, Peddi R, Harrison G, Vandeputte K, Fitzsimmons W, First R, Holman J. Continuing 24 month results of a multicenter, randomized trial comparing three induction agents (alemtuzumab, thymoglobulin and basiliximab) with tacrolimus, mycophenolate mofetil and a rapid steroid withdrawal in renal transplantation. Am J Transplant. 2009;9 Suppl 2:282.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Ciancio G, Burke GW, Gaynor JJ, Roth D, Kupin W, Rosen A, Cordovilla T, Tueros L, Herrada E, Miller J. A randomized trial of thymoglobulin vs. alemtuzumab (with lower dose maintenance immunosuppression) vs. daclizumab in renal transplantation at 24 months of follow-up. Clin Transplant. 2008;22:200-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 72]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
62.  Tan HP, Kaczorowski DJ, Basu A, Unruh M, McCauley J, Wu C, Donaldson J, Dvorchik I, Kayler L, Marcos A. Living donor renal transplantation using alemtuzumab induction and tacrolimus monotherapy. Am J Transplant. 2006;6:2409-2417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
63.  Tan HP, Donaldson J, Basu A, Unruh M, Randhawa P, Sharma V, Morgan C, McCauley J, Wu C, Shah N. Two hundred living donor kidney transplantations under alemtuzumab induction and tacrolimus monotherapy: 3-year follow-up. Am J Transplant. 2009;9:355-366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 52]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
64.  Kaufman DB, Leventhal JR, Axelrod D, Gallon LG, Parker MA, Stuart FP. Alemtuzumab induction and prednisone-free maintenance immunotherapy in kidney transplantation: comparison with basiliximab induction--long-term results. Am J Transplant. 2005;5:2539-2548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 160]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
65.  Hanaway MJ, Woodle ES, Mulgaonkar S, Peddi VR, Kaufman DB, First MR, Croy R, Holman J. Alemtuzumab induction in renal transplantation. N Engl J Med. 2011;364:1909-1919.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Flechner SM, Friend PJ, Brockmann J, Ismail HR, Zilvetti M, Goldfarb D, Modlin C, Mastroianni B, Savas K, Devaney A. Alemtuzumab induction and sirolimus plus mycophenolate mofetil maintenance for CNI and steroid-free kidney transplant immunosuppression. Am J Transplant. 2005;5:3009-3014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 86]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
67.  Hidalgo LG, Campbell PM, Sis B, Einecke G, Mengel M, Chang J, Sellares J, Reeve J, Halloran PF. De novo donor-specific antibody at the time of kidney transplant biopsy associates with microvascular pathology and late graft failure. Am J Transplant. 2009;9:2532-2541.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Alba A, Morales J, Fierro A, Zehnder C, Cao C, Orozco R, Herzog C, Calabrán L, Contreras L, Buckel E. Evaluation of late immunologic parameters among renal transplant recipients induced with Campath-1H. Transplant Proc. 2010;42:253-256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
69.  Vo AA, Lukovsky M, Toyoda M, Wang J, Reinsmoen NL, Lai CH, Peng A, Villicana R, Jordan SC. Rituximab and intravenous immune globulin for desensitization during renal transplantation. N Engl J Med. 2008;359:242-251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 538]  [Cited by in F6Publishing: 494]  [Article Influence: 30.9]  [Reference Citation Analysis (0)]
70.  Takagi T, Ishida H, Shirakawa H, Shimizu T, Tanabe K. Evaluation of low-dose rituximab induction therapy in living related kidney transplantation. Transplantation. 2010;89:1466-1470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
71.  Clatworthy MR, Watson CJ, Plotnek G, Bardsley V, Chaudhry AN, Bradley JA, Smith KG. B-cell-depleting induction therapy and acute cellular rejection. N Engl J Med. 2009;360:2683-2685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 201]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
72.  Tydén G, Genberg H, Tollemar J, Ekberg H, Persson NH, Tufveson G, Wadström J, Gäbel M, Mjörnstedt L. A randomized, doubleblind, placebo-controlled, study of single-dose rituximab as induction in renal transplantation. Transplantation. 2009;87:1325-1329.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Luan FL, Schaubel DE, Zhang H, Jia X, Pelletier SJ, Port FK, Magee JC, Sung RS. Impact of immunosuppressive regimen on survival of kidney transplant recipients with hepatitis C. Transplantation. 2008;85:1601-1606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
74.  Stock PG, Barin B, Murphy B, Hanto D, Diego JM, Light J, Davis C, Blumberg E, Simon D, Subramanian A. Outcomes of kidney transplantation in HIV-infected recipients. N Engl J Med. 2010;363:2004-2014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 402]  [Cited by in F6Publishing: 366]  [Article Influence: 26.1]  [Reference Citation Analysis (0)]
75.  Patel SJ, Knight RJ, Suki WN, Abdellatif A, Duhart BT, Krauss AG, Mannan S, Nezakatgoo N, Osama Gaber A. Rabbit antithymocyte induction and dosing in deceased donor renal transplant recipients over 60 yr of age. Clin Transplant. 2011;25:E250-E256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
76.  Micromedex healthcare series; Greenwood village, CO: Thompson healthcare inclusive, 2011.  .  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Morton RL, Howard K, Webster AC, Wong G, Craig JC. The cost-effectiveness of induction immunosuppression in kidney transplantation. Nephrol Dial Transplant. 2009;24:2258-2269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
78.  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-S155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 666]  [Cited by in F6Publishing: 987]  [Article Influence: 65.8]  [Reference Citation Analysis (0)]