You are on page 1of 6

American Journal of Transplantation 2008; 8: 15–20 C 2008 The Authors

Blackwell Munksgaard Journal compilation 


C 2008 The American Society of

Transplantation and the American Society of Transplant Surgeons


Minireview doi: 10.1111/j.1600-6143.2007.02053.x

Alemtuzumab (Campath-1H) in Kidney Transplantation

G. Ciancio∗ and G. W. Burke III inhibitors (CNIs) (5), the alloimmune component of trans-
plant glomerulopathy (6), and other less well-defined pro-
Department of Surgery, Division of Transplantation, cesses (7). Regimens that can yield similar short-term re-
University of Miami Miller School of Medicine, sults but with less long-term deterioration (8,9) continue to
Miami, FL be sought, perhaps even eventually allowing the develop-
∗ Corresponding author: Gaetano Ciancio, ment of immunologic tolerance (10,11).
gciancio@med.miami.edu
New biologic agents such as (chimeric or humanized) mon-
Kidney transplantation has become the treatment of oclonal antibodies with longer half-lives have been intro-
choice for both the quality of life and survival in pa- duced as induction therapy (2). The resulting prolonged
tients with end-stage renal disease (ESRD). However, biologic effect may allow lower dosages of (nephrotoxic)
the immunosuppressive regimen which allows opti- CNIs, both short- and long-term, and perhaps lower doses
mal kidney transplant outcome remains elusive. One of or avoidance of corticosteroids. One such agent is Alem-
the more promising induction agents, Alemtuzumab, tuzumab, a humanized CD52-specific complement fix-
was introduced to kidney transplantation by Calne in ing (cytotoxic-lymphodepleting) IgG1 monoclonal antibody
the late 1990s with low dose cyclosporine A monother-
first introduced in hemato-oncology by Waldmann and Hale
apy, with the hope of establishing ‘prope’ or near tol-
erance. Subsequent pilot studies with Alemtuzumab (12), then in renal transplantation by Calne (13). The hu-
alone or monotherapy (DSG, Rapa) demonstrated high man CD52 antigen is found in variable concentrations on
rates of acute rejection (AR) along with occasional hu- all peripheral blood mononuclear cells. Alemtuzumab pro-
moral components that lead to abandoning the con- foundly depletes T cells from peripheral blood for several
cept of Alemtuzumab as a ‘magic bullet’ to achieve months with effects, albeit less marked, on B cells, natural
tolerance, prope or otherwise. A number of programs killer cells, and monocytes, (in descending order) and with
(including our own) has since modified maintenance the least effect on CD34+ (immature) hematopoetic stem
immunosuppression using low dose tacrolimus, and cells. (see review 14).
shown acceptable rates of AR, with relatively low inci-
dence of viral infection and lymphoproliferative disor-
Alemtuzumab is currently not indicated for organ transplan-
ders along with cost benefit. However, there are only
three prospective, randomized studies which are small tation and thus patients should be informed of its off-label
with one year or less follow-up, and most published use and its evolving experimental nature.
series utilize historical control groups with relatively
short follow-up. As extrapolation from short-term data
is far from secure, long-term, prospective, randomized Alemtuzumab in Kidney Transplantation
studies with Alemtuzumab will be necessary to deter-
mine the optimal immunosuppressive regimen. Early experience
The Cambridge group performed the first series of human
Key words: Acute rejection, Alemtuzumab, induc- deceased donor renal transplants using Alemtuzumab in-
tion therapy, mycophenolate mofetil, renal allograft, duction therapy as prophylaxis against rejection in com-
sirolimus, tacrolimus
bination with low dose cyclosporine monotherapy (13).
Received 27 July 2007, revised 19 September 2007 and The pilot trial (Table 1) of 31 patients utilized two doses
accepted for publication 28 September 2007 of Alemtuzumab, 20 mg each, given on the day of the
transplant and the following day. Cyclosporine monother-
apy was used posttransplant with trough levels in the range
of 100–150 ng/mL. There were six episodes of steroid-
Introduction responsive rejection with one case of an almost acellular
vasculitic rejection also treated with steroids. With 5-year
Progress in the development of immunosuppressive regi- follow-up (15), the incidence of AR which had been lower
mens has reduced the incidence and severity of acute re- initially in the Alemtuzumab treated group (13), equaled
jection (AR) after renal transplantation. This has resulted that of the control group (contemporaneous kidney trans-
in improved short-term (primarily 1 year) outcomes (1–3), plant recipients all but five of whom were treated with
but with less marked effects on long-term graft survival triple maintenance therapy without induction) and was ap-
(4). This is due in part to the nephrotoxicity of calcineurin proximately 30% due to an increase in late (1–3 years post

15
Ciancio and Burke III

Table 1: Published effects of Alemtuzumab was used in higher doses and without subsequent im-
Yes No Possibly References munosuppression in this report (17), the possibility of
autoimmune phenomena including thyroiditis associated
Tolerance induction
Alemtuzumab alone × 16 with Alemtuzumab, should be recognized.
Alemtuzumab with early DSG × 19
Prope (near) tolerance induction Alemtuzumab with short-term deoxyspergualin
Alemtuzumab with CyA × 13,15 monotherapy: This same group (19) investigated whether
monotherapy Alemtuzumab and deoxyspergualin (DSG) monotherapy
Effective with non-CNI would induce tolerance in humans. Five recipients of
maintenance therapy live donor kidneys were treated perioperatively with
Sirolimus mono therapy × 22,23 Alemtuzumab and DSG in a pilot trial and followed post-
Sirolimus/MMF × 24
operatively without maintenance immunosuppression.
Alemtuzumab serially/MMF × 25
Alemtuzumab was used for peripheral and nodal T-cell
Effective with CNI therapy
Steroid reduction/ ×1 26,29–31, depletion; DSG was chosen for (1) the demonstration
sparing 33–35 of tolerance when added to a T-cell depletion model in
Spaced weaning (Tacro) ×1 27,28 nonhuman primates (20) and for its inhibitory effect on
Effective for treatment of ? 36,37 monocyte/macrophage activation (21). Despite profound
acute rejection T-cell depletion and DSG dosing, all patients developed
Role in future protocols ? reversible rejection that was similar in timing, histology
1 Short-term. and transcriptional profile to that seen in patients treated
CNI = calcineurin inhibitor; DSG = deoxyspergualin; MMF = My- with Alemtuzumab alone (16).
cophenolate Mofetil; Tacro = Tacrolimus.
Answer: No.

kidney transplant) episodes of AR. Furthermore, the serum


creatinine was the same in both groups at 5 years, de- QU: Is Alemtuzumab effective with non-CNI
spite the hope that the lower cyclosporine A concentra- maintenance therapy?
tion in Alemtuzumab recipients would translate to reduced Sirolimus monotherapy: At the University of Wisconsin
nephrotoxicity. Importantly, the Alemtuzumab patients on (22), 29 patients received two doses of 20 mg each of
cyclosporine A monotherapy were ‘no more tolerant than Alemtuzumab on day 0 and day 1. Twenty-four (living donor)
patients on triple therapy’ as judged by graft survival and recipients then received maintenance sirolimus therapy.
incidence of AR (13). Thirty percent in this pilot study experienced significant
rejection episodes, usually between 2–3 weeks postop-
QU: Is Alemtuzumab tolerogenic? eratively, which were typically of a humoral rather than
Alemtuzumab alone: Kirk et al. (16) were the first to use cellular nature. Some were difficult to control, requiring
Alemtuzumab in a pilot trial designed to test the need for a combination of plasmapheresis, cytomegalovirus (CMV)
maintenance immunosuppression in the context of multi- IGm thymoglobulin, rituximab, steroid bolus, mycopheno-
ple doses of Alemtuzumab. Seven nonsensitized recipients late mofetil (MMF) and tacrolimus. Using a modification in
of living donor kidneys received Alemtuzumab without ad- the protocol, the initial dose of Alemtuzumab was given
ditional immunosuppression. Alemtuzumab was adminis- the day before transplantation followed by a dose of thy-
tered at 0.3 mg/kg per dose. Six patients received three moglobulin 1.5 mg/kg on day 1 (patients 26–29). A 2-week
doses in the peritransplant period: four on days –5, –3 course of steroids was also added. Nevertheless, of five
and –1 and two on days –3, –1 and +2. One received patients, three experienced AR.
four doses on days –1, +1, +3 and +5 (11,13). Profound
peripheral lymphocyte and monocyte depletion occurred. At 3-years, the pilot study included a 46% rate of rejec-
Nonetheless, all patients developed rejection episodes tion (humoral and/or cellular). Seven (54%) of these were
within the first month that were characterized by mono- humoral, treated with the regimen described (23). How-
cytic (not lymphocytic) infiltrates with only rare T cells ever, graft and patient survival of 96% and 100%, respec-
in the peripheral blood or allograft. Most were reversed tively, were achieved. Fifty percent of the patients were
with steroid therapy although one required OKT3 as well. maintained on monotherapy without serious infections and
Sirolimus maintenance therapy was then initiated. There there were no malignancies. Because of the high incidence
were no late rejections. of early rejection, however, a brief course of a CNI was rec-
ommended (23).
Of note, one patient developed auto immune thyroid dis-
ease 3 years after transplant/Alemtuzumab induction while Rapa/MMF; no CNI: Flechner et al. (24) in a pilot study
receiving sirolimus monotherapy (17). Autoimmunity has of 22 patients (14 living donor; 8 deceased donors)
also previously been reported in multiple sclerosis patients used Alemtuzumab (30 mg, day 0 and 1) induction
treated with Alemtuzumab (18). Although Alemtuzumab with mycophenolate mofetil (500 mg b.i.d.) and sirolimus

16 American Journal of Transplantation 2008; 8: 15–20


Alemtuzumab

(concentration controlled 8–12 mg/mL) maintenance in or- cophenolate mofetil and higher dose steroids. The Alem-
der to avoid CNI and steroids. With a mean follow-up of tuzumab group overall experienced less rejection than
15.9 months, patient survival was (21/22) 96% and graft the other three groups: Alemtuzumab ∼22%, anti-CD25
survival (19/22) 87%. However, AR occurred in 8 (36.3%- ∼30% and thymoglobulin ∼32% (from figure 2 in Ref.
two humoral). Of 19 surviving grafts, 18 (95%) remain 20) (p = 0.037). The follow-up in the Alemtuzumab group
steroid and 15 (79%) CNI free. Overall infection rates were was 12 months versus 4–6 years for the other groups. A
low, but two patients developed severe acute respiratory subgroup with delayed graft function, treated with Alem-
distress syndrome (ARDS) at month 3 and 7, respectively, tuzumab, experienced less rejection and improved graft
resulting in mortality in one and graft loss in the other. survival statistically p = 0.0096 and 0.0119, respectively,
No cancer or posttransplant lymphoproliferative disease than the control groups. The rate of AR in the Alemtuzumab
(PTLD) was observed. The higher than expected rate of DGF group (n = 23) was 14%, anti-CD25 (n = 127) was
AR, leucopenia and possible pulmonary toxicity raised con- 38% and in the thymoglobulin group (n = 41) 42% (fig-
cerns regarding the efficacy and safety of this protocol. ure 2 in Ref. 20). However, the follow-up was limited to
200 days for Alemtuzumab recipients with DGF (figure 2
Serial Alemtuzumab/MMF; no CNI/steroids: Gruessner in Ref. 20). There was no difference in infection or ma-
et al. (25) reported a nonrandomized study of 75 pancreas– lignancies among the four groups. Further follow-up will
kidney and solitary pancreas recipients who received alem- be necessary to assess outcome in the Alemtuzumab
tuzumab (4 doses for induction and up 12 doses within the group.
first year) and mycophenolate mofetil (≥2 g/day) monother-
apy. Alemtuzumab, 30 mg IV, was given intraoperatively University of Pittsburgh: Reports from the University of
as well as for maintenance dosing. The maximum number Pittsburgh (27) suggested lymphoid depletion with Alem-
of Alemtuzumab doses was limited to 10 within the first tuzumab before rather than after kidney transplantation
year. In a 6-month follow-up, the results were compared would reduce the anticipated donor-specific response and
with a historical group of 266 consecutive pancreas recipi- allow avoidance of high-dose steroids and multiple main-
ents using thymoglobulin induction and tacrolimus mainte- tenance immunosuppressive agents. Three groups were
nance. Pancreas allograft survival at 6 months in the Alem- retrospectively compared: historical controls with no pre-
tuzumab for SPK recipients was 81% (vs. 79%; p ≥ 0.66); treatment (n = 152), thymoglobulin (5 mg/kg) pretreatment
for PAK recipients, 91% (vs. 85%; p ≥ 0.59); and for PTA group (n = 101) and Alemtuzumab (30 mg) pretreatment
recipients, 71% (vs. 84%; p ≥ 0.07). The incidence of a group (n = 90). Patient and graft survival were not signif-
first (reversible) rejection episode at 6 months in the Alem- icantly different in either of the depleted groups versus
tuzumab versus control group for SPK recipients was 41% the historical controls. However, there were markedly dif-
(vs. 9%; p ≥ 0.0003); for PAK recipients, 14% (vs. 10%; ferent incidences and times to AR. In the thymoglobulin-
p ≥ 0.89); and for PTA recipients, 19% (vs. 26%; p ≥ 0.36). pretreated patients, the onset was earlier and the incidence
Based on 6-month data, the combination of alemtuzumab higher (40% at 6 months, ∼50% at 1 year (figure 2 in Ref.
and MMF was associated with a high rejection rate (in SPK 27) (p < 0.001) than in either the Alemtuzumab or histor-
recipients), but good (graft and native) kidney function; it ical controls. The incidence of rejection during the first 6
eliminated undesired CNI and steroid-related side effects. months after Alemtuzumab pretreatment was 1%, how-
Longer follow-up is necessary to evaluate both efficacy and ever, by 12 months the AR rate increased to 20% (figure 2
safety. in Ref. 27). Of note, 3-year graft survival in the thymoglobu-
lin group, which experienced the high rate of AR, was 70%
Answer: No compared to ∼80% in the historical control group (figure 1
in Ref. 27).

Is Alemtuzumab effective with CNI maintenance Spaced weaning of tacrolimus was attempted in 91
therapy? (90.1%) of the 101 recipients of the thymoglobulin pretreat-
University of Wisconsin: After their initial experience with ment group. In 45% of these patients, daily maintenance
Alemtuzumab and sirolimus monotherapy, the Wisconsin therapy was resumed because of AR. In view of the high
group (26) reported a single center retrospective study rate of AR and worse graft survival observed with space
comparing patients who received two doses of Alem- weaning after thymoglobulin pretherapy, spaced wean-
tuzumab at the time of renal transplant in combination ing was delayed for 1 year before being attempted in 83
with a low dose of steroids (10 mg methylprednisolone (91.2%) of the 90 patients of the Alemtuzumab pretreat-
per day), mycophenolate mofetil and either tacrolimus or ment group. With follow-up of 12–18 months, 62 (74%)
cyc1osporine (n = 126) with those who received either an of the 84 Alemtuzumab-treated patients were on spaced
anti-CD25 antibody (n = 799), thymoglobulin (n = 160) or weaning, 14% on daily monotherapy, with only 12%
other antibody treatment (n = 156). The choice of induc- receiving more than a single agent. However, further
tion antibody was made by the operating surgeon based on follow-up of graft survival and change in creatinine is nec-
‘perceived efficacy, side effect profile and cost’ (20). The essary to evaluate the safety and efficacy of Alemtuzumab
latter three groups were in combination with a CNI, my- in this trial.

American Journal of Transplantation 2008; 8: 15–20 17


Ciancio and Burke III

More recently this group has reported results in an LRD groups. The targeted trough level of tacrolimus was be-
kidney transplant protocol with Alemtuzumab and spaced tween 8 and 10 ng/mL for thymoglobulin and daclizumab
weaning of tacrolimus monotherapy (28). The 1-year ac- groups, with a targeted mycophenolate dose of 1 gm
tuarial patient and graft survival was 98.6% and 98.1%, twice daily. In the alemtuzumab treated arm, target
respectively. With larger numbers and better use of im- tacrolimus trough level was lower 4–7 ng/mL (to reduce
munologic monitoring, the 1-year AR episodes were 6.8%. long-term nephrotoxicity,) with 500 mg twice daily doses
They report no CMV or other infection, no DGF and PT DM of mycophenolate and no steroids. AR rates at 1 year
rate of.5% (28). However, longer follow-up will be critical were equivalent, that is 5/30 in all three groups (∼16.6%).
to assess the effectiveness of this approach. In the alemtuzumab group, there was slightly lower GFR
at 1 month, but no difference at 1 year.
The University of Miami: The Miami experience with
Alemtuzumab began with a pilot study in an attempt to In the Alemtuzumab group, 80% remained steroid-free at
(1) reduce both early and long-term CNI nephrotoxicity, 1 year, with lower tacrolimus trough levels and mycophe-
and (2) eliminate steroids. Alemtuzumab (two doses, 30 nolate dosing. There were no differences in other adverse
mg each) was used as induction therapy in 44 de novo re- events. No direct comparison among these three arms is
nal allograft recipients (29). Maintenance Tacrolimus trough possible, because the maintenance regimens are different
levels were 5–7 ng/mL; with reduced MMF dosage (500 (33). However, the efficacy of Alemtuzumab appears simi-
mg twice daily). With a median follow-up of 9 (range 1– lar to that of the other two agents in this small, prospective
19) months, patient and graft survival were both 100%. randomized study. Further follow-up is in progress.
Biopsy-proven AR was diagnosed in four patients (9%).
Four patients (9%) developed infections that required hos- Alemtuzumab versus basiliximab: A nonrandomized,
pitalization. Thirty-eight (86%) remained off long-term cor- retrospective, sequential study comparing outcomes in kid-
ticosteroid therapy. In our experience, the combination of ney transplant recipients induced either with Alemtuzumab
Alemtuzumab, low dose tacrolimus and mycophenolate (n = 123) or basiliximab (n = 155) with a prednisone-free
mofetil, and avoidance of maintenance corticosteroid use maintenance protocol using tacrolimus and mycopheno-
was safe and effective for kidney transplant recipients. late mofetil was reported from Northwestern (34). Mean
follow-up was 33 ± 23 months (range 30–42 months) for
Later reports and our own experience, however, suggested Alemtuzumab and 47 ± 10 months (range 31–65 months)
that a higher incidence of biopsy-demonstrated AR may ex- for the basiliximab group, and ∼2/3 were living donors (33).
ist in the African–American and Hispanic subgroups treated
with Alemtuzumab (30). This remains controversial be- The 1-year actual patient and death censored kidney trans-
cause others, in a small, prospective randomized study, plant survival rates, Alemtuzumab versus basiliximab, were
showed equivalence in AR rates at 1 year comparing Alem- 96.8% versus 99.4%, and 99.2% and 99.4%, respectively
tuzumab to thymoglobulin in an immunologically high-risk (p = n.s.). Although the rate of AR was lower in the Alem-
group that included African–Americans and Hispanics (31). tuzumab group for the first 3 months, by 12 months, the
Of note, the second randomized prospective study com- rate of AR for both groups was equivalent (Alemtuzumab
paring Alemtuzumab with CyA (low dose) monotherapy 14.9%; basiliximab 13.5%). There was no difference in cre-
(n = 20) to triple immunosuppression (CyA, azathioprine atinine concentration at 1 year and minimal change over
and steroids) without induction (n = 10) also reported 36 months. CMV occurred in 21% (Alemtuzumab) and 19%
equivalence (32). However, the study was small with only (basiliximab) in the zero negative recipients, and in both
6-month follow-up, and the AR rate was already relatively groups PTLD occurred in two patients. The Northwestern
high: 25% (Alemtuzumab) versus 20% (standard immuno- team has also demonstrated reduced cost associated with
suppression). alemtuzumab induction (35).

Answer: Yes, but longer follow-up and randomized studies Answer: It is possible to achieve excellent short-term
are necessary to draw meaningful conclusions. (1 year) results with Alemtuzumab. However, comparison
studies suffer from lack of (1) long-term follow-up and (2)
randomized, prospective controlled trials. The best induc-
QU: Is Alemtuzumab better than other induction tion protocol for the long-term remains to be defined.
agents?
Alemtuzumab versus thymoglobulin versus da-
clizumab: Our group (33) subsequently designed the first Alemtuzumab for the Treatment of Acute
randomized prospective trial using three different antibody Rejection in Renal Transplant Patients
induction agents in 90 first renal transplant recipients
from deceased donors: thymoglobulin, Alemtuzumab and There are anecdotal reports describing the use of Alem-
daclizumab. Maintenance immunosuppression included tuzumab in the treatment of AR (36). There are no prospec-
tacrolimus and mycophenolate in all three arms, and tive studies. There appears to be a higher risk of infection
methylprednisolone in the thymoglobulin and daclizumab in patients treated with Alemtuzumab for AR (37).

18 American Journal of Transplantation 2008; 8: 15–20


Alemtuzumab

Alemtuzumab: acute humoral rejection (AHR) in renal of many novel therapeutic agents, from the initial response
transplant recipients of enthusiasm, to a realization that it is not a panacea.
In kidney transplant patients who received Alemtuzumab The learning curve has been through tolerance, prope tol-
induction, AHR appeared to occur with higher frequency, erance, mono/dual agent maintenance therapies to cur-
requiring an aggressive combination of plasmapheresis rent protocols, generally embracing low dose CNIs and
and intravenous immunoglobulin, in addition to steroid ther- steroid sparing successfully. There are suggestions that
apy (19,22,24). One case of AHR with an almost acellular Alemtuzumab is particularly effective in delayed graft func-
vasculitic rejection that was treated with steroids was re- tion, and that there are important cost savings with its use.
ported in Calne’s original study (13). It should be noted that
plasma cells do not express the CD52 epitope in detectable However, preventing AHR will be critically important. It may
concentrations and that memory T (and perhaps B lympho- be necessary to avoid the use of Alemtuzumab in those pa-
cytes) may also be somewhat more refractory to Alem- tients with B-cell positive crossmatches and/or antidonor
tuzumab (38). Kirk et al. (16) reported no AHR, but a high antibody. If the current experience showing low rates of
incidence of reversible AR with monocytic predominance AR, with correspondingly low rates of viral infections, lym-
in the biopsy infiltrates. Notably, chemokine transcript lev- phoproliferative disorders and posttransplant diabetes at
els in these biopsies were consistently high, suggesting 1 year, can be validated in randomized, prospective, long-
the facilitation of infiltrating cells (19). All were respon- term protocols, then Alemtuzumab will assume an impor-
sive to treatment followed by maintenance with steroids tant role in kidney transplantation induction therapy.
or sirolimus or both. Hill et al. (39) reported an early severe
AHR (C4d+) resulting in renal allograft loss in a recipient
treated with Alemtuzumab induction therapy, cyclosporine
Acknowledgment
A, mycophenolate mofetil and steroids. This patient devel-
The authors would like to express their appreciation to Ms. Maruja Chavez
oped donor specific anti-class I antibody. The presence of
for her perspicacious, prescient and perceptive perseverance in the prepa-
antidonor MHC antibodies or B cell crossmatch pretrans- ration of this paper.
plantation is not detailed (39). It is clear that Alemtuzumab
induction, even with additional immunosuppressive agents
does not prevent AHR in renal allograft recipients. References
Alemtuzumab/Tregs 1. Pirsch JD, Miller J, Deierhoi MH, Vicenti F, Filo R. A comparison of
Our group (33) reported significantly more leucopenia with tacrolimus (FK506) and cyclosporine for immunosuppression after
Alemtuzumab than thymoglobulin or daclizumab, but a cadaveric renal transplantation. Transplantation 1997; 63: 977–983.
greater percentage of T regulatory cells as assessed by 2. Ciancio G, Burke GW, Suzart K et al. Daclizumab induction,
number of Fox-P3 mRNA copies, by flow cytometry and tacrolimus, mycophenolate mofetil and steroids as an immunosup-
pression regimen for primary kidney transplant recipients. Trans-
semiquantitative PCR analysis. This was in the context
plantation 2002; 73: 1100–1106.
of low dose mycophenolate mofetil and low dose/level
3. Hariharan S, Johnson CP, Bresnahan BA, Taranto SE, McIntosh
tacrolimus maintenance immunosuppression. However, MJ, Stablein D. Improved graft survival after renal transplantation
Noris et al. (40) using cyclosporine A as the CNI, not in the United States, 1988 to 1996. N Engl J Med 2000; 342: 605–
tacrolimus, did not identify Tregs. In this study, Treg pheno- 612.
type and function was monitored during immune reconsti- 4. Meier-Kriesche HU, Schold JD, Kaplan B. Long-term renal allograft
tution in 21 renal transplant recipients treated with Alem- survival: Have we made significant progress or is it time to rethink
tuzumab and MMF who received sirolimus or cyclosporine our analytic and therapeutic strategies? Am J Transplant 2004; 4:
as maintenance therapy. Three patients, one on sirolimus 1289–1295.
and two on cyclosporine, experienced an AR episode that 5. Nankivell BJ, Borrows RJ, Fung CL, O’Connell PJ, Allen RD, Chap-
man JR. The natural history of chronic allograft nephropathy. N
responded to steroid therapy. In sirolimus treated patients,
Engl J Med 2003; 349: 2326–2333.
hyporesponsiveness demonstrated in vitro was reversed
6. Gloor JM, Sethi S, Stegall MD et al. Transplant glomerulopathy:
by Treg depletion. T cells from cyclosporine treated pa- Subclinical incidence and association with alloantibody. Am J Trans-
tients were anergic. Thus, in their study CD4+CD25 high plant 2007; 7: 2124–2132.
Treg expansion in renal transplant patients, appeared to 7. Naesens M, Lerut E, Damme BV, Vanrenterghem Y, Kuypers DRJ.
occur with sirolimus but not CyA maintenance immuno- Tacrolimus exposure and evolution of renal allograft histology in
suppression. Translating this observation into a real clini- the first year after transplantation. Am J Transplant 2007; 7: 2114–
cal benefit (41), because sirolimus protocols without CNI 2123.
have shown higher rates of AR, remains a challenge for the 8. Ciancio G, Burke GW, Gaynor JJ et al. A randomized long-term trial
future. of tacrolimus/sirolimus vs. tacrolimus /mycophenolate mofetil vs.
cyclosporine (Neoral® )/sirolimus in renal transplantation. I. Drug
interactions and rejection at one year. Transplantation 2004; 77:
Conclusion 244–251.
9. Ciancio G, Burke GW, Gaynor JJ et al. A randomized long-term
The use of Alemtuzumab as induction therapy in kidney trial of tacrolimus/sirolimus vs. tacrolimus/mycophenolate mofetil
transplantation, has evolved over the past decade, typical vs. cyclosporine (Neoral® )/sirolimus in renal transplantation. II.

American Journal of Transplantation 2008; 8: 15–20 19


Ciancio and Burke III

Survival, function, and protocol compliance at one year. Transplan- 26. Knechtle SJ, Fernandez LA, Pirsch JD et al. Alemtuzumab in renal
tation 2004; 77: 252–258. transplantation: The University of Wisconsin experience. Surgery
10. Ciancio G, Miller J, Garcia-Morales RO et al. Six year clinical ef- 2004; 136: 754–760.
fect of donor bone marrow infusions in renal transplant patients. 27. Shapiro R, Basu A, Tzan H et al. Kidney transplantation under min-
Transplantation 2001; 71: 827–835. imal immunosuppression after pretransplant lymphoid depletion
11. Spitzer TR, Delmonico F, Tolkoff-Rubin N et al. Combined histo- with thymoglobulin or Alemtuzumab. J Am Coll Surg 2005; 200:
compatibility leukocyte antigen-matched donor bone marrow and 505–515.
renal transplantation for multiple myeloma with end stage renal 28. Tan HP, Kaczorowski DJ, Basu M et al. Living donor renal transplan-
disease: The induction of allograft tolerance through mixed lym- tation using Alemtuzumab induction and tacrolimus monotherapy.
phohematopoietic chimerism. Transplantation 1999; 68: 480–484. Am J Transplant 2006; 6: 2409–2417.
12. Waldmann H, Hale G. Alemtuzumab: From concept to clinic. 29. Ciancio G, Burke GW, Gaynor JJ et al. The use of Alemtuzumab
Philos Trans R Soc Lond B Biol Sci 2005; 360: 1707– as induction therapy in renal transplantation: Preliminary results.
1711. Transplantation 2004; 78: 426–433.
13. Calne R, Moffatt SD, Friend PJ et al. Alemtuzumab 1H allows low- 30. Ciancio G, Sageshima J, Gaynor JJ et al. The use of Alemtuzumab
dose cyclosporine monotherapy in 31 cadaveric renal allograft re- as induction therapy to achieve steroid avoidance in African–
cipients. Transplantation 1999; 68: 613–616. American and Hispanic first renal transplant recipients [abstract].
14. Flynn JM, Byrd JC. Alemtuzumab monoclonal antibody therapy. Am J Transplant 2006; 1324: 513.
Curr Opin Oncol 2000; 12: 574–581. 31. Thomas PG, Woodside KJ, Lappin JA, Vaidya S, Rajaraman S,
15. Watson CJE, Bradley JA, Friend PJ et al. Alemtuzumab Gugliuzza KK. Alemtuzumab (Alemtuzumab 1H) induction with
(Alemtuzumab 1H) induction therapy in cadaveric kidney tacrolimus monotherapy is safe for high immunological risk renal
transplantation—efficacy and safety at five years. Am J Transplant transplantation. Transplantation 2007; 83: 1509–1512.
2005; 5: 1347–1353. 32. Vathsala A, Ona ET, Tan SY et al. Randomized trial of Alemtuzumab
16. Kirk AD, Hale DA, Mannon RB et al. Results from a human renal for prevention of graft rejection and preservation of renal func-
allograft tolerance trial evaluating the humanized CD52-specific tion after kidney transplantation. Transplantation 2005; 80: 765–
monoclonal antibody alemtuzumab (Alemtuzumab). Transplanta- 774.
tion 2003; 76: 120–129. 33. Ciancio G, Burke G, Gaynor JJ et al. A randomized trial of three renal
17. Kirk AD, Hale DA, Swanson SJ, Mannon RB. Autoimmune thyroid transplant induction antibodies: Early comparison of tacrolimus,
disease after renal transplantation using depletional induction with mycophenolate mofetil, and steroid dosing, and newer immune-
Alemtuzumab. Am J Transplant 2006; 6: 1084–1085. monitoring. Transplantation 2005; 80: 457–465.
18. Coles AJ, Wing M, Smith S et al. Pulsed monoclonal antibody 34. Kaufman DB, Leventhal JR, Axelrod D et al. Alemtuzumab in-
treatment and autoimmune thyroid disease in multiple sclerosis. duction and prednisone-free maintenance immunotherapy in kid-
Lancet 1999; 354: 1691–1695. ney transplantation: Comparison with basiliximab induction—long-
19. Kirk AD, Mannon RB, Kleiner DE et al. Results from a human re- term results. Am J Transplant 2005; 5: 2539–2548.
nal allograft tolerance trial evaluating T-cell depletion with alem- 35. Kaufman DB, Leventhal JR, Gallon LG, Parker MA. Alemtuzumab
tuzumab combined with deoxypergualin. Transplantation 2005; 80: induction and prednisone-free maintenance immunotherapy in si-
1051–1059. multaneous pancreas-kidney transplantation comparison with rab-
20. Thomas JM, Contreras JL, Jiang XL. Peritransplant tolerance in- bit antithymocyte globulin induction—long-term results. Am J
duction in macaques: Early events reflecting the unique synergy Transplant 2006; 6: 331–339.
between immunotoxin and deoxyspergualin. Transplantation 1999; 36. Thomas PG, Ishihara K, Vaidya S, Gugliuzza KK. Alemtuzumab and
68: 1660. renal transplant rejection. Clin Transplant 2004; 8: 759—761.
21. Amemiya H. Deoxyspergualin: Immunosuppressive properties. 37. Peleg AY, Husain S, Kwak EJ et al. Opportunistic infections in 547
Norwalk, CY: Appleton and Lange, 1989. organ transplant recipients receiving Alemtuzumab, a humanized
22. Knechtle SJ, Pirsch JD, Fechner J Jr et al. Alemtuzumab induction monoclonal CD-52 antibody. Clin Infect Dis 2007; 44: 204–212.
plus rapamycin monotherapy for renal transplantation: Results of 38. Pearl JP, Parris J, Hale DA et al. Immunocompetent T-cells with
a pilot study. Am J Transplant 2003; 3: 722–730. memory-like phenotype are the dominant cell type following
23. Barth RN, Janus CA, Lillesand CA et al. Outcomes at 3 years of antibody-mediated T-cell depletion. Am J Transplant 2005; 5: 465–
prospective pilot study of Alemtuzumab and sirolimus immuno- 474.
suppression for renal transplantation. Transplant Int 2006; 19: 885– 39. Hill P, Galiardini E, Ruggenenti P, Remuzzi G. Severe early acute
892. humoral rejection resulting in allograft loss in renal transplant recip-
24. Flechner SM, Friend PJ, Brockmann J et al. Alemtuzumab induc- ient with Alemtuzumab induction therapy. Nephrol Dial Transplant
tion and sirolimus plus mycophenolate mofetil maintenance for 2005: 1741–1744.
CNI and steroid-free kidney transplant immunosuppression. Am J 40. Noris M, Casiraghi F, Todeschini M et al. Regulatory T cells and
Transplant 2005; 5: 3009–3014. T cell depletion: Role of immunosuppressive drugs. J Am Soc
25. Gruessner RWG, Kandaswamy R, Humar A, Gruessner AC, Suther- Nephrol 2007; 18: 1007–1018.
land DER. Calcineurin inhibitor and steroid-free immunosuppres- 41. Kang SM, Tang Q, Bluestone JA. CD4+CD25+ regulatory T cells in
sion in pancreas-kidney and solitary pancreas transplantation. transplantation: Progress, challenges and prospects. Am J Trans-
Transplantation 2005; 79: 1184–1189. plant 2007; 7: 1457–1463.

20 American Journal of Transplantation 2008; 8: 15–20

You might also like