Submit manuscript...
eISSN: 2378-3176

Urology & Nephrology Open Access Journal

Review Article Volume 3 Issue 6

Induction agent in low immunological risk; the Indian scenario

Pradeep Shenoy,1,2 Julie M Bridson,2 Ahmed Halawa,2,4 Ajay Sharma2,3

1KS Hegde Medical Academy, India
2Faculty of Health and Life Science, Institute of Learning and Teaching, University of Liverpool, UK
3Royal Liverpool University Hospitals, UK
4Sheffield Teaching Hospitals, UK

Correspondence: Ajay Sharma, Consultant Transplant Surgeon and Director of Core Surgical Training, Northwest of England Deanery, Associate Director of Post Graduate Courses in Transplantation and Honorary Senior Lecturer, University of Liverpool Link 9C, Royal Liverpool University Hospital Liverpool, L78XP, UK

Received: April 08, 2016 | Published: December 12, 2016

Citation: Shenoy P, Bridson JM, Halawa A, et al. Induction agent in low immunological risk; the Indian scenario. Urol Nephrol Open Access J. 2016;3(6):193-199. DOI: 10.15406/unoaj.2016.03.00106

Download PDF

Abstract

Chronic kidney disease (CKD) is a global health problem because of increasing prevalence, high cost of management and risk factor for ischemic heart disease. Renal transplantation remains the current best form of therapy for end stage renal disease (ESRD). Renal transplantation with living kidney donor is better compared to deceased donor transplantation in terms of survival and quality of life. Use of induction agents in renal transplantation has evolved over the years from initial use of muromonab, anti-thymocyte globulins (ATG), and IL-2 blockers to alemtuzumab. Multiple studies have compared these agents in different risk groups and scenarios. There is strong evidence for using ATG compared to IL-2 blockers in high risk group, however, the same does not apply to low risk group. There is a paucity of data regarding the use of induction agents in the Indian sub-continent. The role of induction agents in this population which is prone to opportunistic infections, side effects of drugs and where cost of treatment takes a priority, are not well known especially in low immunological risk group. This review article summarizes the role of various induction agents in different risk groups with relevance to Indian population.

Keywords: Induction; ATG; Basiliximab; Renal; Transplantation and low risk; India; Population

Abbreviations

CKD: Chronic Kidney Disease; ESRD: End Stage Renal Disease; ATG: Anti-Thymocyte Globulins; RRT: Renal Replacement Therapy; CAPD: Continuous Ambulatory Peritoneal Dialysis; CNI: Calcineurin inhibitors; ATG: Anti-thymocyte Globulin; rATG: Rabbit Anti-thymocyte Globulin; ALG: Anti-lymphocyte Globulin; ARDS: Acute Respiratory Distress Syndrome; CMV: Cytomegalovirus; BPAR: Biopsy Proven Acute Rejection; MMF: Mycophenolate Mofetil

Introduction

India faces an uphill task of providing affordable tertiary health care to its citizens due to lack of public infrastructure. The crude age adjusted incidence rate of end stage renal disease (ESRD) in India is 151 per million population [1,2]. According to the Indian CKD registry, about 74.5% of patients do not receive any form of renal replacement therapy (RRT) and only 2.5% receive renal transplantation. It has been estimated that about 90% of ESRD patients die within months of diagnosis in south Asia as patients cannot afford such treatments due to poverty [2].

ESRD patient’s have2 options: maintenance dialysis (haemodialysis or peritoneal dialysis) or renal transplantation. Those patients who are fit enough, renal transplantation are the best option when compared to dialysis in regards to patient outcomes and cost. A recent study (2015) done by Diego Rosseli [3] concluded that renal transplantation, from a living or deceased donor, improves the overall survival rate and quality of life and is a cost saving alternative compared with dialysis. Renal transplantation living or deceased donor improves both life expectancy and quality of life for the recipient and can be done with living or deceased donor. Patient and graft survival is better by a transplant from a living donor than from a deceased donor. It is estimated that in India 15,000 patients commence maintenance haemodialysis, 3000 new patients choose continuous ambulatory peritoneal dialysis (CAPD) and 3500 patients undergo renal transplantation every year [4]. It is estimated that in India 150,000 people are waiting for renal transplantation and 90% of patients in waiting list die without getting an organ transplant [1].

The corner stone of renal transplantation is the use of immunosuppression to modify the alloimmune response to ‘the foreign body’ i.e. ‘renal allograft. Since the time of first successful human renal transplantation among twins in 1954 there have been lots of advances in the field of use of immunosuppressant. The era of immunosuppressant began (1960s) with the use of steroids followed by use of combination of steroids and azathioprine. Introduction of calcineurin inhibitors (CNI) and induction agents (depleting and non-depleting) have remarkably decreased the rate of acute rejection however with little effect on long term out comes. (Table 1) lists the immunosuppressant medication used in renal transplantation

Induction agents

ATG (anti-thymocyte globulin)

Basiliximab

Daclizumab

Alemtuzumab

Rituximab

Maintenance agents

Corticosteroids

Mycophenolate mofetil (MMF)

Azathioprine

Tacrolimus

Cyclosporine

Belatacept

Sirolimus

Table 1: The list of immunosuppressive medication used in renal transplant.

Induction drugs 

Induction agents are immunosuppressive drugs which are given during the peri-transplant period with the intention of decreasing the rate of acute rejection. The choice of induction regimen varies from transplant centre to centre and individual to individual. Initial induction agents in 1990s were OKT3 and horse ATG, both of which have decreased significantly over the years. Commonly used induction agents are equine ATG and IL-2 receptor blockers. They can be classified as T cell depleting and T cell non-depleting. T cell depleting agents are most commonly used followed by basiliximab according to US data 2009 (Table 2-4).

Generic

Brand

Classification

FDA indication

FDA Approval

Manufacturer

Basiliximab

Simulect

IL-2 receptor antibody, monoclonal antibody, CD25

Prevention of rejection in kidney and liver transplant

1998

Novartis

Daclizumab

Zenapax

IL-2 receptor antibody, monoclonal antibody, CD25

Prevention of rejection in kidney transplant

1997-2009

Roche

Rabbit ATG

Thymoglobulin

Polyclonal anti-T cell

Treatment of acute rejection in kidney transplantation

1998

Genzyme-sanofi

Horse ATG

ATGAM

Polyclonal anti- T cell

Treatment of acute rejection in kidney transplantation
Aplastic anaemia

1981

Pfizer

Alemtuzumab

Campath

Monoclonal antibody, CD52

Treatment of B cell CLL

2001-2012

Berlex laboratories

Muromonab, OKT3

Orthoclone

Monoclonal antibody, CD3

Treatment of acute rejection in liver, heart, kidney transplantation.

1986-2009

Janssen-cilag

Table 2: lists a variety of induction agents used in renal transplantation.

Agents

Example

1.T cell depleting

Anti-thymocyte globulin (ATG)

2.T cell non depleting

IL-2 receptor inhibitor e.g. basiliximab

Table 3: Classification of induction agents.

 

Atgam

Lymphoglobulin

Thymoglobulin

ATG-Fresenius

Manufacture

Pfizer

Various

Genzyme-sanofi

Fresenius-biotech, GmBH

Location

New York, USA

Cambridge, USA

Grafelfing, Germany

Species

Horse

Horse

Rabbit

Rabbit

Concentration (mg/ml)

50

10-20

5

20

Immunogen

Human thymus

Human thymus

Human thymus

Jurkat lymphoblastic cell line

Dosage per day (mg/kg)

10-30

10

1.25-2.5

1-5

Table 4: Immunoglobulin’s available for induction in renal transplantation.

Muromonab

Muromonab (OKT3) is a monoclonal antibody derived from mice which binds to CD3 receptor. It was initially used for induction. It is given in a dose of 5-10 mg/day for about 10-14 days with cumulative dose of 70mg. Common side effect include cytokine release syndrome with fever, chills, nausea and vomiting. Cytokine release may lead to pulmonary edema and hemodynamic instability that is more likely in those who have fluid overload. Few patients have developed anti-mouse antibodies following muromonab infusion. Consequently, Muromonab is rarely used nowadays for induction in renal transplant.

Anti-thymocyte globulin

Anti-thymocyte globulin (ATG) is produced by immunizing animals with human thymus derived lymphoid cells. They are commercially available in 4 forms, with rabbit anti-thymocyte globulin (rATG) being more efficacious, hence it is the preferred choice over equine anti-lymphocyte globulin (ALG). ATG is not FDA approved for induction in renal transplantation; however, it is used rather commonly in North America. Various regimens use daily intravenous preparation for 5 to 10 days either for induction or for treatment of steroid resistant rejection usual dosage is 1.5 mg/kg for 3-5 days [5-14].

High cumulative dose of ATG and long duration of treatment is associated with opportunistic infection and malignancy, however, lower dose (<3mg/kg) may be insufficient to prevent acute rejection [15]. ATG contains multiple antibodies which bind to variety of cells in the body including red blood cells, white blood cells, endothelium and platelets. ATG is one of the most potent immunosuppressive agents available and can suppress lymphocytes for up to 24 hours. Being non-specific, ATG leads to marked immunosuppressive effect that predisposes to infection and malignancy. Most common side effects are infusion reactions including fever, chills, lymphocytopenia, pancytopenia and rarely acute respiratory distress syndrome (ARDS). Cytopenias require dose adjustment and dose of ATG can be modified based on CD2 and CD3 counts. Peripheral administration of ATG can lead to venous thrombosis which can be reduced by concomitant infusion of heparin and hydrocortisone [16,17].

Alemtuzumab

Alemtuzumab or Campath-1H is a monoclonal antibody against CD 52. CD 52 is present on most of the lymphocytes B and T cells and monocytes but not on hematopoietic precursor cells. It’s not FDA approved for induction and is used as off label drug. It suppresses lymphocytes for many months to years and is given in a dose of 30 mg or 0.3mg/kg IV in a single dose, or sometimes two doses based on lymphocyte count [18]. Common side effects include cytokine release syndrome as can be seen with ATG, however to a lesser extent. Its use has been associated with infectious complications and autoimmune thyroiditis and has been used to treat acute rejection in post transplant scenario with poor outcomes. The initial use of alemtuzumab in renal transplant recipients was associated with intense and prolonged lymphocyte depletion, increased late antibody-mediated graft rejection and increased rates of serious infection [19-22].

IL-2 Inhibitors

IL-2 is highly expressed in activated T cells. IL-2 inhibitors (basiliximab and daclizumab) bind specifically to its alpha subunit (CD25) and block further activation of T cells. Basiliximab is a chimeric antibody which can suppress T cell for up to 25 to 35 days. It issued for induction regimen only. Normally it is given in a dose of 20 mg on the day of transplantation and day 4 of post transplantation. No major side effects have been reported with basiliximab. It’s the only induction drug which is FDA approved

Evidence for various induction regimens

United Network for Organ Sharing (UNOS) registry data showed that induction with any agent is better than no induction therapy at all [23].

Basiliximab or no induction

Major trials on basiliximab have proved its efficacy in preventing acute cellular rejection with maintenance of cyclosporine, steroids with or without antimetabolites [24,25]. Other trials have shown improving trends in rejection rate though not statistically significant [26]. Another retrospective study from SRTR (scientific renal transplant registry) showed small but significant reduction in acute rejection with basiliximab compared to no induction [27]. However, none of these trials showed any difference in graft survival or patient survival. One meta-analysis published in 2004, which included 14 trials enrolling 2410 patients compared basiliximab with placebo and showed reduced acute rejection rate at one year, but the incidence of graft loss was similar [28].

ATG compared to no induction

In deceased donor transplantation, two randomized control trials have shown reduced acute rejection rate with rATG. In one study (2001), combination of rATG and tacrolimus was used whilst the other study (2003) used rATG and cyclosporine. Both studies showed reduced rejection rate but with higher incidence of leucopoenia, thrombocytopenia and opportunistic infection especially cytomegalovirus (CMV). Although the incidence of acute rejection was lower in rATG group compared to placebo, there was no difference in graft or patient survival [29,30].

Meta-analysis by Liu Y which included six randomized control trials with 823 patients did not show any difference between ATG versus basiliximab in outcomes that included biopsy proven acute rejection (BPAR), graft survival and patient survival [31]. One of the drawbacks of this meta-analysis was the heterogeneous study population and it included all preparations of ATG. One of the studies included in the meta-analysis used equine ATG or basiliximab with delayed and early initiation of cyclosporine respectively, had similar rejection rate [32].In another multicenter randomized study in Europe (2002) with rATG alone or basiliximab with steroids, mycophenolate mofetil (MMF), early cyclosporine (basiliximab group) and delayed cyclosporine (rATG group), there was no difference in BPAR or renal function at one year [33]. Long term follow up at 5 years did not show any difference in outcome including incidence of CMV infection. Limitation of this study was sub optimal immunosuppression, low immunological risk group and lack of data on pre-transplant CMV seropositive status.

 In a randomized study Mourad G from Europe (2004) comparing rATG and basiliximab where cyclosporine was introduced when creatinine was less than 2.7 mg/dl, and already commenced on with steroids and MMF, did not show any difference in rejection rate at one year (basiliximab 9.6%, ATG group 9.4%) [34]. There was a statistically significant increase in leucopenia (19.2% vs 51.0%, p=0.0007), CMV infection rate (21.2% vs 41.5%, p=0.025) in rATG group despite chemoprophylaxis for CMV in seropositive donor/seronegative recipient transplantation.

In another large trial (2006) involving patients at high risk for acute rejection or delayed graft function, who received renal transplant from deceased donor, induction therapy consisting of ATG as compared with basiliximab had reduced incidence and severity of acute rejection but not the incidence of delayed graft function. Positively, even at 5 year follow-up there was a decreased incidence of rejection in the rATG group [35]; however this group had higher incidence of leucopoenia although the rate of CMV infection was similar.

Alemtuzumab versus other induction drugs

In one large multicentre randomized control trial published in 2011, which compared alemtuzumab versus basiliximab (low immunological risk) and alemtuzumab versus rATG (high immunological risk), the incidence of BPAR was less in the alemtuzumab group (in low immunological risk) although it did not translate into better graft survival [36]. Alemtuzumab group had a higher incidence of late acute rejection compared to other two induction agents. A meta-analysis (2012) on alemtuzumab as induction agent which include six randomized trials with 808 patients showed reduced incidence of BPAR in the alemtuzumab group compared to other induction group (except in high risk recipients), however, it did not translate into better graft or patient survival [37]. High risk recipients as defined by panel reactive antibody (PRA) >10%, re-transplant, black race had similar BPAR in all the groups

Immunological risk

Immunological risk of a particular recipient depends upon both donor characters and recipient status. The principle of induction is to minimize the risk of rejection while keeping the risk of side effects such as cytopenia, infection and malignancy as low as possible. The choice of induction agent depends upon the immunological risk status of recipient. Table 5 shows characteristic of low and high risk groups.

Lower risk

High risk

Zero HLA match

HLA mismatch

Live donors

Younger recipient and older donor age

Caucasian ethnicity

African-American ethnicity

Low panel reactive antibody (PRA)

High PRA

Absence of donor specific antibody (DSA)

Presence of DSA

Blood group compatibility

Blood group incompatibility

Immediate graft function

Delayed graft function

Short cold ischemia time

Long cold ischemia time

First transplant

Re-transplant.

Table 5: Characteristics of low and high risk groups.

Live donors

A retrospective study compared induction versus no induction in living donor transplantation in two time periods (1998-2002) versus (2003-2008), which showed no difference in graft and patient outcome at five year follow up; however the type of induction agent was not analysed [38]. Induction therapy with different drugs has shown short term benefit in the form of reduced incidence and severity of acute rejection without any effect on long term graft and patient survival. Some studies have compared no induction with individual induction agents such as ATG, basiliximab and alemtuzumab in live kidney transplant with different results. A single centre study compared rATG with no induction control group showed superior graft and patient outcome at 5 years [39]. Similar results have been found in single centre studies in unrelated renal transplantation [40]. Other studies have found lower acute rejection rate at one year with alemtuzumab and basiliximab compared to controls with no induction [41,42].

A prospective study from Egypt (2008) showed26% reduction in incidence of acute rejection at one year with basiliximab compared to no induction with maintenance immunosuppression of steroids, cyclosporine and azathioprine [43]. Various Induction agents have been used with early corticosteroid withdrawal. A prospective multicentre randomised control trial (2010) with early corticosteroid withdrawal with ATG induction versus chronic corticosteroid therapy without induction , showed similar BPAR at 6 and 12 months in both groups, however, early corticosteroid withdrawal group had better lipid profile and less weight gain [44].

Deceased donor

Like live donor renal transplantation cadaveric renal transplantation with zero mismatches are considered low immunological risk. A review showed that in zero mismatched cadaveric renal transplantation, various induction agents (ATG, basiliximab, alemtuzumab) had lower rejection rates at 6 months post transplantation compared to no induction, although both groups had similar patient and graft survival [45].

Standard criteria donor

A secondary analysis of a randomized control trial (2006) comparing ATG with basiliximab in standard criteria deceased donor transplant showed reduced incidence of acute rejection, graft loss or death in the ATG group [46].

Delayed graft function

A prospective study in deceased donor transplantation showed that an intra-operative first dose of rATG is better than post operative ATG with respect to renal function at day 14 post transplantation. Some of the beneficial effects of intra-operative ATG could be due to prevention of acute rejection and, thereby, preventing delayed graft function [15].However, there are studies which do not support the hypothesis that ATG prevents delayed graft function [35].

Ethnicity

The African American race is at higher immunological risk of rejection when compared to Caucasian. A retrospective study in the US showed that there was no difference in outcome in terms of graft and patients survival in African American and Caucasian with ATG or Basiliximab induction. However, some other studies in Chinese patients did not show beneficial effect of basiliximab induction in preventing acute rejection [47,48].

Recipient age

In contrast to young kidney transplant recipients, elderly recipients have a high incidence of post operative complications including infection due to low immunity. A retrospective database analysis in deceased donor transplantation in elderly recipients showed that ATG was preferable in high risk recipient (PRA>20%, black race, re-transplant) with high risk donors (ECD, DCD, cold ischemia time >24 hours). ATG was also preferred in low risk recipients with high risk donors, although this study was not a randomized control trial and was blighted with multiple bias [49].

Immunosuppressant minimization

There are studies which justify the use of ATG in low immunological risk groups who have undergone early corticosteroid withdrawal. In a randomized control trial by Vanrenterghem et al. [50] ATG was shown to be effective in preventing acute rejection compared to no induction in early corticosteroid withdrawal group. Additionally, in a retrospective study of live donor recipients with early corticosteroid withdrawal, induction with r ATG was superior to no induction with respect to lower incidence of acute rejection [51].

Locally, in St John’s Medical College Hospital, Bangalore, we perform live donor renal transplantation without any induction in low immunological risk group. The decision to avoid induction in low immunological risk group is driven by cost factors and absence of long term benefit in Indian population. We commence immunosuppression two days prior to the date of transplantation and the recipient receives bolus IV steroids intra-operatively followed by a tapering dose of oral steroids, tacrolimus and MMF post-op. In patients with lower risk of rejection, the avoidance of induction therapy decreases the cost of transplantation, which is the most important determining factor for opting for renal transplantation as a modality of renal replacement therapy. Our local centre protocol for induction in low immunological risk living donor renal transplant (Table 6).

Day of transplant

Drugs used

Comments

-2 day of transplant

Start tacrolimus and MMF

Tacrolimus- 0.1mg/kg
MMF- 30mg/kg in two divided doses

Transplant day

Inj methylprednisolone 1000mg
No induction

 

Day 1

Tab prednisolone 20 mg+ Tacrolimus+ MMF

 

Day 5

Tacrolimus +MMF+ steroids

Monitor Tacrolimus level
Target trough 12-15mcg/ml

4 weeks post transplant

Minimize prednisolone by tapering 2.5 mg/week
+ Tacrolimus +MMF

AUC for MMF is not done.

6 months

Prednisolone 5mg + Tacrolimus+ MMF

Target Tacrolimus level 8-12 mcg/ml

1 year

Prednisolone 5 mg+ Tacrolimus+ MMF

Target Tacrolimus level 5-8 mcg/ml

Table 6: Transplant protocol at our local centre.

Evidence for induction from Indian studies

There are limited numbers of studies from India which compare different induction regimens. Data is even more sparse when it comes to low immunological risk groups. An observational study by Gundlapalli S et al. (52) intermediate risk group with poor HLA match, compared basiliximab induction with placebo in 46 living donor renal transplant recipients with tacrolimus, MMF and steroids as the maintenance immunosuppressive regimen; there was no difference in acute rejection rate. The study concluded that expensive induction regimen of basiliximab in the intermediate immunological risk group did not confer any added advantage to existing triple immunosuppression. The limitations of the study were lack of randomization, small number of patients and short follow up.

A study by Mahendra A et al. [53] in living donor kidney transplant recipients with long term follow up showed a reduced rejection rate with basiliximab compared to no induction; however, there was no effect on graft survival or patient survival at five years. The occurrence of infection was also higher in basiliximab group compared to placebo. Similar results have been found in other major studies elsewhere in world [28,43]. In a retrospective study that compared ATG and basiliximab in 86 living donor renal transplantation recipients, there was no difference in long-term outcomes including patient and graft survival [54]. There are no randomized control trials from India which compared various induction agents with placebo in low immunological risk groups. The long term outcome of avoiding of induction is also unknown in this population.

One of the drawbacks of avoiding induction agents in low immunological risk groups is the requirement for intensive follow up post transplantation as theoretically these patients are at higher risk of rejection. In the era of tacrolimus, MMF and steroid combination, the avoidance of induction is worth the benefit in low immunological risk groups especially in a country like India where financial and physical resources are the major limiting factors. In addition, there is a lower risk of opportunistic infection with avoidance of Induction. Treatment of opportunistic infection with antimicrobials adds to post transplantation expenditure. However, the final word on the optimal induction regimen in low immunological risk group in Indian setting is yet to be established and there is clear need for well conducted randomised trials by investigators in India.

Conclusion

The immunological risk profile of the recipients determines the choice of induction agents. In high immunological risk status, ATG has been shown to improve graft outcomes, whereas in low immunological risk group basiliximab and ATG are equally efficacious, although with higher incidence of side effects with ATG. In low immunological risk groups the evidence supporting the use of induction drugs is rather weak, especially from India, as there no well conducted randomized studies. In addition, the infectious complications of induction regimen and cost of chemoprophylaxis with certain induction agents adds cost to existing transplant expenditure. In the background of no proven long term benefit of using induction agents in recipients with low immunological risk in Indian population, the avoidance of induction agents may be a good option in order to save the cost and potential morbidity.

References

  1. Modi GK, Jha V (2006) The incidence of end-stage renal disease in India: a population-based study. Kidney Int 70(12): 2131-2133.
  2. Abraham G (2008) The challenges of renal replacement therapy in Asia. Nat Clin Pract Nephrol 4(12): 643.
  3. Rosselli D, Rueda JD, Diaz CE (2015) Cost effectiveness of kidney transplantation compared with chronic dialysis in end stage renal disease. Saudi J Kidney Dis Transpl 26(4): 733-738.
  4. Agarwal SK, Srivastava RK (2009) Chronic kidney disease in India: Challenges and solution. Nephron Clin Pract 111(3): c197-203.
  5. Brennan DC, Flavin K, Lowell JA, Howard TK, Shenoy S, et al. (1999) A randomized, double- blinded comparison of thymoglobulin versus Atgam for induction immunosuppressive therapy in adult renal transplant recipients. Transplantation 67(7): 1011-1018.
  6. Agha IA, Rueda J, Alvarez A, Singer GG, Miller BW, et al. (2002) Short course induction immunosuppression with thymoglobulin for renal transplant recipients. Transplantation 73(3): 473-475.
  7. Gurk-Turner C, Airee R, Philosophe B, Kukuruga D, Drachenberg C, et al. (2008) Thymoglobulin dose optimization for induction therapy in high risk kidney transplant recipients. Transplantation 85(10): 1425-1430.
  8. Hardinger KL, Rasu RS, Skelton R, Miller BW, Brennan DC (2010) Thymoglobulin induction dosing strategies in a low-risk kidney transplant population: three or four days? J Transplant 957549.
  9. Klem P, Cooper JE, Weiss AS, Gralla J, Owen P, et al. (2009) Reduced dose rabbit anti-thymocyte globulin induction for prevention of acute rejection in high-risk kidney transplant recipients. Transplantation 88(7): 891-896.
  10. Laftavi MR, Alnimri M, Weber-Shrikant E, Kohli R, Said M, et al. (2011) Low-dose rabbit antithymocyte globulin versus basiliximab induction therapy in low-risk renal transplant recipients: 8-year follow- up. Transplant 43(2): 458-461.
  11. Peddi VR, Bryant M, Roy-Chaudhury P, Woodle ES, First MR (2002) Safety, efficacy, and cost analysis of thymoglobulin induction therapy with intermittent dosing based on CD3+ lymphocyte counts in kidney and kidney- pancreas transplant recipients. Transplantation 73(9): 1514-1518.
  12. Starzl TE, Murase N, Abu-Elmagd K, Gray EA, Shapiro R, et al. (2003) Tolerogenic immunosuppression for organ transplantation. Lancet 361(9368): 1502-1510.
  13. Stevens RB, Mercer DF, Grant WJ, Freifeld AG, Lane JT, et al. (2008) Randomized trial of single-dose versus divided-dose rabbit anti-thymocyte globulin induction in renal transplantation: an interim report. Transplantation 85(10): 1391-1399.
  14. Wong W, Agrawal N, Pascual M, Anderson DC, Hirsch HH, et al. (2006) Comparison of two dosages of thymoglobulin used as a short-course for induction in kidney transplantation. TransplInt 19(8): 629-635.
  15. Goggins WC, Pascual MA, Powelson JA, Magee C, Tolkoff-Rubin N, et al. (2003) A prospective, randomized, clinical trial of intraoperative versus postoperative Thymoglobulin in adult cadaveric renal transplant recipients. Transplantation 76(5): 798-802.
  16. Erickson AL, Roberts K, Malek SK, Chandraker AK, Tullius SG, et al. (2010) Analysis of infusion-site reactions in renal transplant recipients receiving peripherally administered rabbit antithymocyte globulin as compared with basiliximab. TransplInt 23(6): 636-640.
  17. Wiland AM, Fink JC, Philosophe B, Farney AC, Schweitzer EJ, et al. (2001) Peripheral administration of thymoglobulin for induction therapy in pancreas transplantation. Transplant Proc 33(1-2): 1910.
  18. Calne R, Moffatt SD, Friend PJ, Jamieson NV, Bradley JA, et al. (1999) Campath IH allows low-dose cyclosporine monotherapy in 31 cadaveric renal allograft recipients. Transplantation 68(10): 1613-1616.
  19. Mourad G, Garrigue V, Squifflet JP, Besse T, Berthoux F, et al. (2001) Induction versus non induction in renal transplant recipients with tacrolimus based immunosuppression. Transplantation 72(6): 1050-1055.
  20. Knechtle SJ, Pirsch JD, H Fechner JJ, Becker BN, Friedl A, et al. (2003) Campath-1H induction plus rapamycin monotherapy for renal transplantation: results of a pilot study. Am J Transplant 3(6): 722-730.
  21. Martin SI, Marty FM, Fiumara K, Treon SP, Gribben JG, et al. (2006) Infectious complications associated with alemtuzumab use for lymphoproliferative disorders. Clin Infect Dis 43(1): 16-24.
  22. Peleg AY, Husain S, Kwak EJ, Silveira FP, Ndirangu M, et al. (2007) Opportunistic infections in 547 organ transplant recipients receiving alemtuzumab, a humanized monoclonal CD-52 antibody. Clin Infect Dis 44(2): 204-212.
  23. Cai J, Terasaki PI (2010) Induction immunosuppression improves long-term graft and patient outcome in organ transplantation: an analysis of United Network for Organ Sharing registry data. Transplantation 90(12): 1511-1515.
  24. Kahan BD, Rajagopalan PR, Hall M (1999) 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 67(2): 276-284.
  25. Nashan B, Moore R, Amlot P, Schmidt AG, Abeywickrama K, et al. (1997) Randomised trial of basiliximab versus placebo for control of acute cellular rejection in renal allograft recipients. CHIB 201 International study group. Lancet 350(9086): 1193-1198.
  26. Lawen JG, Davies EA, Mourad G, Oppenheimer F, Molina MG, et al. (2003) 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 75(1): 37-43.
  27. Gralla J, Wiseman AC (2010) The impact of IL2ra induction therapy in kidney transplantation using tacrolimus- and mycophenolate- based immunosuppression. Transplantation 90(6): 639-644.
  28. Webster AC, Playford EG, Higgins G, Chapman JR, Craig JC (2004) Interleukin 2 receptor antagonists for renal transplant recipients: a meta-analysis of randomized trials. Transplantation 77(2): 166-176.
  29. Mourad G, Garrigue V, Squifflet JP, Besse T, Berthoux F, et al. (2001) Induction versus noninduction in renal transplant recipients with tacrolimus based immunosuppression. Transplantation 72(6): 1050-1055.
  30. Charpentier B, Rostaing L, Berthoux F, Lang P, Civati G, et al. (2003) 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 75(6): 844-851.
  31. Liu Y, Zhou P, Han M, Xue CB, Hu XP, et al. (2010) Basiliximab or antithymocyte globulin for induction therapy in kidney transplantation: a meta-analysis. Transplant Proc 42(5): 1667-1670.
  32. Sollinger H, Kaplan B, Pescovitz MD, Philosophe B, Roza A, et al. (2001) Basiliximab versus antithymocyte globulin for prevention of acute renal allograft rejection. Transplantation 72(12): 1915-1919.
  33. Lebranchu Y, Bridoux F, Buchler M, Le Meur Y, Etienne I, et al. (2002) Immunoprophylaxis with basiliximab compared with antithymocyte globulin in renal transplant patients receiving MMF-containing triple therapy. Am J Transplant 2(1): 48-56.
  34. Mourad G, Rostaing L, Legendre C, Garrigue V, Thervet E, et al. (2004) Sequential protocols using basiliximab versus antithymocyte globulins in renal-transplant patients receiving mycophenolatemofetil and steroids. Transplantation 78(4): 584-590.
  35. Brennan DC, Daller JA, Lake KD, Cibrik D, Del Castillo D, et al. (2006) Rabbit antithymocyte globulin versus basiliximab in renal transplantation. N Engl J Med 355(19): 1967-1977.
  36. Hanaway MJ, Woodle ES, Mulgaonkar S, Peddi VR, Kaufman DB, et al. (2011) Alemtuzumab induction in renal transplantation. N Engl J Med 364(20): 1909-1919.
  37. Zhang X, Huang H, Han S, Fu S, Wang L (2012) Alemtuzumab induction in renal transplantation: a meta-analysis and systemic review. Transpl Immunol 27(2-3): 63-68.
  38. Emami S, Huang E, Kuo HT, Kamgar M, Bunnapradist S (2012) Multivariate analysis of antibody induction therapy and their associated outcomes in live donor kidney transplantation in the recent era. Clin Transplant 26(2): 351-358.
  39. Hardinger KL, Schnitzler MA, Koch MJ, Labile E, Stirnemann PM, et al. (2006) Thymoglobulin induction is safe and effective in live-donor renal transplantation: a single centre experience. Transplantation 81(9): 1285-1289.
  40. Miller JT, Collins CD, Stuckey LJ, Luan FL, Englesbe MJ, et al. (2009) Clinical and economic outcomes of rabbit antithymocyte globulin induction in adults who received kidney transplants from living unrelated donors and received cyclosporine-based immunosuppression. Pharmacotherapy 29(10): 1166-1174.
  41. Schenker P, Ozturk A, Vonend O, Krüger B, Jazra M, et al. (2011) Single-dose thymoglobulin induction in living-donor renal transplantation. Ann Transplant 16(2): 50-58.
  42. Lim WH, Chang SH, Chadban SJ, Campbell SB, Dent H, et al. (2009) Interleukin-2 receptor antibody reduces rejection rates and graft loss in live-donor kidney transplant recipients. Transplantation 88(10): 1208-1213.
  43. Sheashaa HA, Bakr MA, Ismail AM, Mahmoud KM, Sobh MA, et al. (2008) Basiliximab induction therapy for live donor kidney transplantation: a long-term follow-up of prospective randomized controlled study. Clin Exp Nephrol 12(5): 376-381.
  44. Woodle ES, Peddi VR, Tomlanovich S, Mulgaonkar S, Kuo PC, et al. (2010) A prospective, randomized, multicenter study evaluating early corticosteroid withdrawal with Thymoglobulin in living donor kidney transplantation. Clin Transplant 24(1): 73-83.
  45. Kuo HT, Huang E, Emami S, Pham PT, Wilkinson AH, et al. (2012) Effects of antibody induction on transplant outcomes in human leukocyte antigen zero-mismatch deceased donor kidney recipients. Transplantation 93(5): 493-502.
  46. Hardinger KL, Brennan DC, Schnitzler MA (2009) Rabbit antithymocyte globulin is more beneficial in standard kidney than in extended donor recipients. Transplantation 87(9): 1372-1376.
  47. Jindal RM, Das NP, Neff RT, Hurst FP, Falta EM, et al. (2009) Outcomes in African- Americans vs. Caucasians using thymoglobulin or interleukin- 2 receptor inhibitor induction: analysis of USRDS database. Am J Nephrol 29(6): 501-508.
  48. Lo YC, Ho HC, Wu MJ, Chen CH, Cheng CH, et al. (2012) Interleukin-2 receptor antagonist does not decrease biopsy-proven acute rejection among adult Chinese kidney transplant recipients. Ren Fail 34(7): 856-861.
  49. Gill J, Sampaio M, Gill JS, Dong J, Kuo HT, et al. (2011) Induction immunosuppressive therapy in the elderly kidney transplant recipient in the United States. Clin J Am Soc Nephrol 6(5): 1168-1178.
  50. Vanrenterghem Y, Lebranchu Y, Hene R, Oppenheimer F, Ekberg H (2000) Double-blind comparison of two corticosteroid regimens plus mycophenolate mofetil and cyclosporine for prevention of acute renal allograft rejection. Transplantation 70(9): 1352-1359.
  51. Martin ST, Roberts KL, Malek SK, Tullius SG, Vadivel N, et al. (2011) Induction treatment with rabbit antithymocyte globulin versus basiliximab in renal transplant recipients with planned early steroid withdrawal. Pharmacotherapy 31(6): 566-573.
  52. Gundlapalli S, Rathi M, Kohli HS, Jha V, Sharma A, et al. (2013) Efficacy of basiliximab induction in poorly matched living donor renal transplantation. Indian J Nephrol 23(6): 409-412.
  53. Atlani M, Sharma RK, Gupta A (2013) Basiliximab induction in renal transplantation: long term outcomes. Saudi J Kidney Dis Transpl 24(3): 473-479.
  54. Kesiraju S, Paritala P, Rao Ch UM, Athmakuri SM, Reddy VS, et al. anti-thymocyte globulin versus basiliximab induction in renal transplant recipients: Long term outcomes. Saudi J Kidney Dis Transpl 25(1): 9-15.
Creative Commons Attribution License

©2016 Shenoy, et al. This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.