Sie sind auf Seite 1von 16

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/23997631

ChemInform Abstract: Infectious Complications


of Antilymphocyte Therapies in Solid Organ
Transplantation

ARTICLE in CLINICAL INFECTIOUS DISEASES MARCH 2009


Impact Factor: 8.89 DOI: 10.1086/597089 Source: PubMed

CITATIONS READS

70 24

2 AUTHORS, INCLUDING:

Jay A Fishman
Massachusetts General Hospital
238 PUBLICATIONS 10,675 CITATIONS

SEE PROFILE

Available from: Jay A Fishman


Retrieved on: 06 April 2016
IMMUNOCOMPROMISED HOSTS INVITED ARTICLE
David R. Snydman, Section Editor

Infectious Complications of Antilymphocyte Therapies


in Solid Organ Transplantation
Nicolas C. Issa and Jay A. Fishman
Infectious Disease Division, Massachusetts General Hospital, Harvard Medical School, Boston

Antilymphocyte therapies are widely used for immunosuppression in solid organ transplantation. These agents have varied
mechanisms of action, with resulting differences in the intensity and duration of immunosuppression and in associated
infectious complications. Induction therapy with antithymocyte globulins is associated with a greater incidence of cytomeg-
alovirus, Epstein-Barr virus, and BK polyomavirus infections, compared with therapy with interleukin (IL)2a receptor
antagonists. However, long-term experience with the IL-2a receptor antagonists is lacking. By contrast, the treatment of graft
rejection with T celldepleting antibodies is associated with an increased risk of opportunistic infections. This is likely a

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


reflection of the intensification of immunosuppression in the treatment of graft rejection and, often, a failure to link the use
of antilymphocyte agents to prophylaxis for infection. The use of T celldepleting agents, especially in the treatment of acute
graft rejection, must be linked to monitoring and risk-adjusted prophylaxis for Pneumocystis, other fungi, Epstein-Barr virus,
BK polyomavirus, and cytomegalovirus infection.

Increasing rates of infection after solid organ transplantation against IL-2a receptor (IL-2R; anti-CD25); and alemtuzumab,
have been attributed to use of antilymphocyte induction ther- a pan-T celldepleting anti-CD52 monoclonal antibody. Use
apies and intensification of regimens for maintenance immu- of OKT3 (muromonab-CD3), a murine monoclonal, T cell
nosuppression [1]. Antilymphocyte therapies are widely used depleting antibody is less common. The T celldepleting agents
in organ transplantation for induction therapy and treatment are also used in the treatment of acute graft rejection. Other
of graft rejection. Each agent has distinct mechanisms of action, agents are being investigated for use in transplantation, in-
with differences in the intensity and duration of immunosup- cluding antibodies to recombinant human leukocyte-associated
pression. Infections commonly associated with these agents ne- functional antigen 1, antibody-associated immunotoxins, and
cessitate the development of appropriate preventative strategies. fusion proteins of human leukocyte-associated functional an-
Induction therapy blocks T cell activation and antigen rec- tigen 3 with immunoglobulin fragments.
ognition at the time of transplantation, particularly in recipients Most trials of antilymphocyte therapies have focused on graft
at high immunologic risk of graft rejection. This strategy min- rejection rates, graft function, and other noninfectious out-
imizes exposure to nephrotoxic calcineurin inhibitors and re- comes. Reporting of specific infections is generally limited to
duces the use of corticosteroids and other maintenance ther- cytomegalovirus (CMV), Epstein-Barr virus (EBV), and BK
apies [2]. The most frequently used induction agents include polyomavirus (BKV) infections, with few data regarding specific
polyclonal antithymocyte globulin (ATG); T celldepleting syndromes or pathogens.
agents from rabbit (rATG) or horse (hATG; lymphocyte im-
munoglobulin [equine antithymocyte globulin]); basiliximab ATG
and daclizumab, non-T celldepleting monoclonal antibodies
The most commonly used ATGs are polyclonal antibodies pre-
pared from sera from rabbits or horses that have been im-
Received 18 July 2008; accepted 5 November 2008; electronically published 10 February munized with thymocytes or T cell lines. rATG and hATG are
2009.
Reprints or correspondence: Dr. Jay A. Fishman, Transplant Infectious Disease and
commonly used in the United States, whereas ATG-Fresenius
Compromised Host Program, 55 Fruit St., GRJ 504, Boston, MA 02114 (jfishman@partners.org). prepared from rabbits injected with Jurkat cellsis used ex-
Clinical Infectious Diseases 2009; 48:77286 tensively in Europe. ATGs are not interchangeable [37]. All
 2009 by the Infectious Diseases Society of America. All rights reserved.
1058-4838/2009/4806-0014$15.00
ATGs provide dose-dependent depletion of T cells. Effects on
DOI: 10.1086/597089 T cell depletion and risk of infection depend on the dosing

772 CID 2009:48 (15 March) IMMUNOCOMPROMISED HOSTS


strategy (i.e., total dose and duration). Typical ATG induction of acute graft rejection with ATG in liver transplant recipients
therapy takes 35 days (range, 110 days) [811]. Longer was not associated with increased risk of bacterial infections,
courses are used to avoid calcineurin inhibitorinduced compared with that risk for patients with graft rejection treated
nephrotoxicity. with steroids or patients who did not experience graft rejection
rATG has a half-life of 30 days [12]. In addition to T cell [37].
depletion, rATG induces apoptosis of B cell lineages and in- CMV. The association between antithymocyte therapies
terferes with dendritic cell, regulatory T cell, and natural killer and CMV infection is well established [17, 24, 3842]. Fever
cell functions [13]. Although levels of rATG decrease by 1 and the release of TNFa after ATG administration stimulate
month after initiation of treatment, significant long-term def- cellular nuclear factor kB and viral replication via nuclear factor
icits in T cell reconstitution that have been linked to impaired kB binding to the promoter region of the CMV immediate-
thymopoiesis often persist beyond 1 year [9, 14, 15]. CD3+ cell early antigen gene [43, 44]. Additional factors in CMV acti-
suppression is less intense and of shorter duration after hATG vation may include the depletion of T helper cells, inversion
administration than after rATG administration, with absolute of the CD4/CD8 ratio, and shifts toward Th2 cytokines [3, 45].
lymphopenia resolving within 14 days after treatment initiation The timing and incidence of CMV infection after ATG induc-
[9]. ATG-Fresenius (9 mg/kg/day for 5 doses) induces rapid T tion therapy depends on the type and dosage of ATG, donor
cell depletion that persists for up to 1 year after treatment, with and recipient CMV serologic status before transplantation, and
gradual recovery to pretransplantation levels by day 730 [14]. whether CMV prophylaxis was given (tables 1 and 2). Most
The development of neutralizing antibodies against xenogeneic trials have included asymptomatic infection (i.e., infection de-
immunoglobulin may limit the efficacy of subsequent courses tectable by microbiologic assay) with CMV syndrome (fever
of ATG. Administration of ATG is often associated with syn- and neutropenia) and tissue-invasive disease in their criteria
dromes including fever and chills and, occasionally, hypoten- for CMV infection. In most prospective, randomized trials

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


sion, chest pains, serum sickness, congestive heart failure, pul- comparing ATG with other induction agents, the incidence of
monary edema, leukopenia, thrombocytopenia, eosinophilia, or CMV infection was not increased when induction therapy was
rash. coupled with adequate CMV prophylaxis [18, 2528, 31]. In a
study without CMV prophylaxis, the rate of CMV infection
ATG and the Risk of Infection was higher in the rATG group than it was in the basiliximab
Bacterial infection. The impact of ATG on bacterial infec- group (38% vs. 11.7%; P p .005) [17]. In one study in which
tions is unclear. Multiple cofactors are generally present, in- CMV infection was more common with daclizumab therapy
cluding technical complications from surgery, urinary and vas- than with hATG therapy, more recipients at high risk of in-
cular catheters, and complex immunosuppressive regimens. fection (i.e., CMV-seronegative recipients of organs from se-
However, bacterial infections are the most common form of ropositive donors) received daclizumab therapy [29].
infection reported after rATG induction therapy (table 1) [16 In general, antigraft-rejection therapy with ATG without
23]. In kidney and kidney-pancreas transplantation, urinary CMV prophylaxis is associated with an increased rate and se-
tract infections are most common, followed by wound infec- verity of CMV infection in renal transplant recipients [47].
tions [9, 1619, 21, 24]. Bacteremia, sepsis, and pneumonia Inflammation associated with acute graft rejection may con-
have also been reported [1719, 32, 33]. Enterobacteriaceae, tribute to CMV reactivation during immunosuppression [47].
most often Escherichia coli and Enterococcus species, are the In simultaneous pancreas-kidney transplant recipients, treat-
most-frequently isolated uropathogens [16]. Nocardia infec- ment of graft rejection with rATG was associated with earlier
tions have been reported in 3 lung transplant recipients (in- and more-severe CMV infection, compared with treatment with
fections were due to Nocardia nova, Nocardia farcinica, and ATG-Fresenius or daclizumab induction [40]. In a study of
Nocardia asteroides complex) after induction therapy with renal transplant recipients, a higher rate of CMV infection as-
rATG, despite prophylaxis with trimethoprim-sulfamethoxa- sociated with basiliximab therapy was attributed to greater use
zole; these infections reflect profound immunosuppression, an- of concomitant cytolytic therapy (ATG or OKT3) among pa-
timicrobial resistance, or inadequate dosing [34]. In most pro- tients receiving basiliximab than among patients receiving rATG
spective trials, ATG was not associated with an increased risk (17.5% vs. 7.8%; P p .02) [21].
of bacterial infection, compared with that risk with no induc- EBV and EBV-induced posttransplantation lymphoprolife-
tion therapy [24] or other induction therapies (e.g., IL-2R an- rative disorder (PTLD). The incidence of malignancy in gen-
tagonists and alemtuzumab) (table 1) [17, 18, 26, 28, 29, 31]. eral, and PTLDs in particular, is increased among organ trans-
Sepsis and multiple renal allograft abscesses (due to Pseudo- plant recipients, compared with that in the general population
monas aeruginosa) have been reported after rATG treatment [4857]. In most cases, PTLD is caused by the transformation
for acute graft rejection [35, 36]. In a small series, treatment of B lymphocytes by EBV [5864]. In EBV-seronegative trans-

IMMUNOCOMPROMISED HOSTS CID 2009:48 (15 March) 773


Table 1. Risk of cytomegalovirus (CMV) and other infections in randomized, prospective trials, by induction therapy.

Maintenance CMV serostatus CMV infection, Other infections,


Study, Duration of immunosuppression Antithymocyte D+/R, no. (%) no. (%) of no. (%) of
treatment group Transplant type follow-up therapy CMV prophylaxis agent of patients P patients P patients P

Mourad et al. [24] (n p 309)


rATG (n p 151) Kidney 12 months Tacrolimus, ACV or GCV in- rATG, 1.25 mg/kg 21 (13.9) NS 49 (32.5) .009 UTI, 47 (31.1); UTI, NS; HSV,
azathioprine, steroids duction, 51%; 10 days HSV, 27 (17.9) .001
no induction,
31.6%
No rATG (n p 158) Kidney 12 months Tacrolimus, ACV or GCV in- None 25 (15.8) 30 (19.0) UTI, 52 (32.9);
azathioprine, steroids duction, 51%; HSV, 9 (5.7)
no induction,
31.6%
Hartwig et al. [25] (n p 44)
rATG (n p 22) Lung 8 years CsA, azathioprine, steroids IV GCV, twice rATG, 1.5 mg/kg Excluded 13 (59) .55 Non-CMV serious 1.999
daily 2 3 days infections, 13
weeks, fol- (59)
lowed by 4
times daily 2
weeks
No rATG (n p 22) Lung 8 years CsA, azathioprine, steroids IV GCV twice None Excluded 10 (45) Non-CMV serious
daily 2 infections, 12

774
weeks, fol- (55)
lowed by 4
times daily 2
weeks
Hardinger et al. [7] (n p 72)
Thymoglobulin (n p 48) Kidney 5 years CsA, azathioprine, steroids Oral GCV, 1 g 3 Thymoglobulin, 8 (17) NS 6 (13) .056
times daily 1.5 mg/kg 7
36 months days
L-IG (n p 24) Kidney 5 years CsA, azathioprine, steroids Oral GCV, 1 g 3 L-IG, 15 mg/kg 5 (21) 8 (33)
times daily 7 days
36 months
Lebranchu et al. [17] (n p 103)
rATG (n p 50) Kidney 6 months CsA, MMF, steroids None rATG, 1.01.5 11 (22) NS 19 (38) .005 Bacterial, 48 (96); Bacterial, NS; vi-
mg/kg/day viral, 39 (78); ral, NS; fungal,
610 days fungal, 2 (4) NS
Basiliximab (n p 51) Kidney 6 months CsA, MMF, steroids None Basiliximab, 20 9 (18) 6 (11.7) Bacterial, 37 (72);
mg on day 0 viral, 21 (41);
and day 4 fungal, 1 (2)
Brennan et al. [21] (n p 278)
rATG (n p 141) Kidney 12 months CsA, MMF, steroids Oral or IV GCV rATG, 1.5 mg/kg/ 21 (14.9) NS 11 (7.8) .02 UTI, 55 (39); con- UTI, .04; con-
14 days, fol- day 5 days firmed bacte- firmed bacte-
lowed by oral rial, 74 (52.5); rial, .01; non-
GCV 3 non-CMV viral, CMV viral, .04;
months 30 (21.3); fun- fungal, 1.999
gal, 20 (14.2)

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


Basiliximab (n p 137) Kidney 12 months CsA, MMF, steroids Oral or IV GCV Basiliximab, 20 31 (22.6) 24 (17.5) UTI, 37 (27); con-
14 days, fol- mg on day 0 firmed bacte-
lowed by oral and day 4 rial, 51 (37.2);
GCV 3 non-CMV viral,
months 16 (11.7); fun-
gal, 20 (14.6)
Mattei et al. [26] (n p 80)
rATG (n p 42) Cardiac 6 months CsA, MMF, steroids For CMV D+/R, rATG, 2.5 mg/kg/ 6 (14.3) .238 10 (23.8) NS Overall, 19 (45.2) .366
oral GCV or day 35
VGCV 3 days
months; other,
per local
practice
Basiliximab (n p 38) Cardiac 6 months CsA, MMF, steroids For CMV D+/R, Basiliximab, 20 11 (28.9) 6 (15.8) Overall, 13 (34.2)
oral GCV or mg on day 0
VGCV 3 and day 4
months; other,
per local
practice
Abou-Ayache et al. [27] (n p
115)
rATG (n p 57) Kidney 12 months CsA, MMF, steroids Oral GCV 1 g 3 rATG, 11.5 mg/ 18 (33) NS 28 (51) NS
times daily kg/day 49
3.5 months doses; ad-
justed to CD2/
CD3 count !20
cells/mm3
Daclizumab (n p 58) Kidney 12 months CsA, MMF, steroids Oral GCV 1 g 3 Daclizumab, 1 19 (35) 21 (39)
times daily mg/kg/day ev-
3.5 months ery 2 weeks

775
5 doses
Sollinger et al. [28] (n p 138)
L-IG (n p 65) Kidney 12 months CsA, MMF, steroids Inconsistent L-IG, 15 mg/kg/ 11 (17) NS Overall, 50 (77) NS
day 14 days
Basiliximab (n p 70) Kidney 12 months CsA, MMF, steroids Inconsistent (1 Basiliximab, 20 13 (19) Overall, 53 (76)
center; no pro- mg on day 0
phylaxis for 21 and day 4)
patients in bas-
iliximab group)
Mullen et al. [29] (n p 50)
L-IG (n p 25) Lung 12 months CsA or tacrolimus, MMF, GCV or VGCV L-IG, 10 mg/kg/ 1 (4) .05 4 (24) .03 Overall, 20 (80) .7
steroids day 58
days
Daclizumab (n, 25) Lung 12 months CsA or tacrolimus, MMF, GCV or VGCV Daclizumab, 2 7 (28) 12 (72) Overall, 22 (88)
steroids mg/kg on day
0, then 1 mg/
kg on day 4
Hershberger et al. [30] (n p
434)
Daclizumab (n p 216) Cardiac 12 months CsA, MMF, steroids GCV 510 mg/kg/ Daclizumab, 1 47 (21.8) .77 11 (5.1) NS Severe, 15 (6.9) NS
day 2 mg/kg 5
weeks, then doses
follow local
protocol

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


No daclizumab (n p 218 Cardiac 12 months CsA, MMF, steroids GCV 510 mg/kg/ None 50 (22.9) 12 (5.8) Severe, 16 (7.7)
day 2
weeks, then
follow local
protocol
Ashcraft et al. [18] (n p 130)
rATG (n p 66) Kidney and 9 months Tacrolimus, MMF, steroids Oral VGCV 36 rATG, alternate 10 (15) 4 (6) NS Overall, 35 (53); Overall, .08; fun-
pancreas months days fungal/viral, 12 gal/viral, NS
(18)
Alemtuzumab (n p 64) Kidney and 9 months Tacrolimus, MMF, steroids Oral VGCV 36 Alemtuzumab, 30 15 (23) 4 (6.25) Overall, 24 (38);
pancreas months mg once fungal/viral, 8
(13)
Ciancio et al. [31] (n p 90)
rATG (n p 30) Kidney 15 months Tacrolimus, MMF, steroids IV GCV 3 days, rATG, 1 mg/kg/ 1 (3) .36 Overall, 7 (23) .6
followed by day 7 days
oral VGCV 3
months
Alemtuzumab (n p 30) Kidney 15 months Tacrolimus, MMF, steroids IV GCV 3 days, Alemtuzumab, 0 (0) Overall, 6 (20)
followed by 0.3 mg/kg on
oral VGCV 3 day 0 and day
months 4
Daclizumab (n p 30) Kidney 15 months Tacrolimus, MMF, steroids IV GCV 3 days, Daclizumab, 1 0 (0) Overall, 4 (13)
followed by mg/kg/day on
oral VGCV 3 day 0 and ev-
months ery 2 weeks
4 doses

NOTE. CMV infection includes asymptomatic CMV infection (antigenemia, PCR detection), CMV syndrome, and CMV disease. ACV, acyclovir; ATG, antithymocyte globulin; CsA, cyclosporine A; D+/R, donor
seropositive and recipient seronegative; GCV, ganciclovir; IV, intravenous; L-IG, lymphocyte immunoglobulin; MMF, mycophenolate mofetil; NS, not significant (exact P value not reported); rATG, rabbit ATG; VGCV,
valganciclovir.

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


plant recipients, the risk of PTLD is increased by coinfection therapy in the absence of prophylaxis [1719, 21, 26, 28, 29,
with CMV and increased intensity of immunosuppression [60, 31, 32]. In most randomized studies, the risk of fungal infection
65]. Induction and graft-rejection therapies using various an- after ATG therapy was similar among different ATG agents
tilymphocyte globulins have been associated with an increased (table 1). False-positive assay results for Histoplasma antigens
risk of PTLD (table 3) [6671]. Not all studies support this may occur in patients receiving rATG therapy [86, 87]. In a
association (table 3) [7072]; many of these studies are affected retrospective, multivariate analysis of renal transplant recipi-
by variability in risk factors between patients, including vari- ents, the rate of invasive fungal infection was not increased by
ability in the type and dosage of induction therapy, type of ATG induction, whereas graft rejection and graft-rejection ther-
organs transplanted, maintenance of immunosuppression, EBV apy increased the risk of invasive fungal infection [88].
serostatus of donor and recipient, and duration of follow-up.
The highest risk of PTLD is in EBV-seronegative recipients of OKT3 (MUROMONAB-CD3)
organs from seropositive donors, particularly children, who OKT3 is a T celldepleting mouse monoclonal antibody that
require long-term monitoring, especially after intensification of has been associated with an increased rate of severe infection
immunosuppression. (notably CMV reactivation [8991] and fungal infection [88])
BKV. Evidence is emerging that links ATG induction ther- and an increased risk of PTLD [67, 70, 92, 93]. The frequency
apy to increased rates of BKV viremia and BKV-associated ne- of OKT3 use has decreased because of the availability of agents
phropathy. BKV-associated nephropathy is an infectious injury with fewer first-doserelated adverse effects (i.e., fever, rigors,
to renal allografts that affects 1%10% of kidney transplant chest pain, dyspnea, hypotension, nausea, vomiting, and di-
recipients and is associated with the intensity of immunosup- arrhea). Patients who receive OKT3 commonly develop anti-
pression [76]. Some studies link ATG induction therapy to the idiotypic antibodies that block efficacy with subsequent ad-
risk of BKV viremia and BKV-associated nephropathy [7780]. ministration [94].

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


rATG induction therapy has been found to be an independent
risk factor for BKV-associated nephropathy, as were tacrolimus- IL-2R ANTAGONISTS (ANTI-CD25)
mycophenolate mophetil and corticosteroid maintenance ther-
IL-2R antagonists (e.g., basiliximab and daclizumab) are mono-
apy [77, 78]. In a prospective trial, BKV viremia occurred in
clonal antibodies that target CD25, the a-chain of the receptor
19.4% of renal transplant recipients at 6 months and was as-
that is expressed on the surface of early progenitors of T and
sociated with higher induction doses of rATG [80]. In a ret- B cells and by activated mature T and B lymphocytes; resting
rospective cohort, induction with polyclonal antibodies was lymphocytes are not targeted. CD25 participates in lymphocyte
associated with an increased rate of BKV-associated nephropa- differentiation, activation, and proliferation. These agents are
thy [79]. The use of antilymphocyte globulins for the treatment used for induction therapy and in maintenance immunosup-
of steroid-resistant graft rejection has been associated with in- pression. Because of the relatively recent introduction of these
creased BKV replication in patients also receiving tacrolimus- agents, clinical experience remains limited but suggests that the
or mycophenylate mophetilbased regimens. Use of pulsed- number of infectious complications may be reduced, compared
dose corticosteroids is a major risk factor for BKV-associated with that for other induction therapies. These agents are not
nephropathy [81]. generally used for the treatment of acute graft rejection.
Hepatitis C Virus (HCV). The use of induction therapy in Basiliximab. Basiliximab is a chimeric murine-human
HCV-infected organ transplant recipients, especially in liver monoclonal antibody that binds selectively to the high-affinity
recipients, may increase HCV replication and accelerate pro- IL-2R [9597]. Basiliximab is generally administered within 2
gression to cirrhosis. This progression is accelerated by CMV h before transplantation and 4 days after transplantation. Bas-
coinfection. Our experience demonstrates accelerated HCV iliximab has a long half-life (13.4 days in adults and 9.4 days
replication with rATG induction therapy for liver transplan- in children) [98, 99]. Induction therapy achieves complete IL-
tation but no increased rate of hepatic graft injury, especially 2R saturation and suppression for 46 weeks in adult kidney
in patients who tolerate antiviral therapy. HCV infection is transplant recipients [96, 97, 99]. In pediatric kidney trans-
associated with increased mortality in kidney transplant recip- plantation, the mean duration of saturation is 42 days [100].
ients [82, 83]; however, antiT cell therapy (rATG or basilix- Daclizumab. Daclizumab is a humanized IL-2R antagonist
imab) was not associated with accelerated viral infection, cir- that contains the hypervariable region of the murine antibody
rhosis, or increased mortality in HCV-seropositive renal against the IL-2R a-subunit [95]. Daclizumab is administered
transplant recipients [84, 85]. every 2 weeks for 5 doses. The half-life of Daclizumab is 20
Fungal infection. Pneumonia due to Pneumocystis species days [101]. The dose used for induction therapy saturates the
and other invasive fungal infections have been reported in solid- IL-2R on circulating lymphocytes for at least 3 months after
organ transplant patients who have received rATG induction transplantation [101].

IMMUNOCOMPROMISED HOSTS CID 2009:48 (15 March) 777


Table 2. Induction therapies and mean time to onset of cytomegalovirus (CMV) infection.

Time to onset
Type of Maintenance CMV serology D+/R, Incidence of CMV infection, of CMV infection,
Study, induction therapy transplant immunosuppression no. (%) of patients CMV prophylaxis % of patients median days (range)
Abou-Ayache [27]
rATG (n p 55) Kidney CsA, MMF, steroids 18 (33) D+/R, universal prophylaxis with GCV 3.5 51 (infection, syndrome, and disease) 29 (1689)
months; D+/R+ or D/R+, preemptive
therapy
Daclizumab (n p 54) Kidney CsA, MMF, steroids 19 (35) D+/R, universal prophylaxis with GCV 3.5 39 (CMV infection, syndrome, and 57 (29168)
months; D+/R+ or D/R+, preemptive disease)
therapy
Huurman [40]
ATG-Fresenius (n p 19) Pancreas-kidney CsA, MMF, steroids 5 (26.3) Universal prophylaxis with GCV 34 months 52.6 (viremia) 114.5 (34180)
Daclizumab (n p 20) Pancreas-kidney CsA, MMF, steroids 4 (20) Universal prophylaxis with GCV 34 months 60 (viremia) 159.5 (139180)
Peleg et al. [46]: alemtuzumab (n p 547) Any Tacrolimus monotherapy Unknown Universal prophylaxis 6 months for kidney, 26 (CMV disease) 85 (7254)
pancreas, lung, intestinal, multivisceral; pre-
emptive therapy for liver recipients

NOTE. ATG, antithymocyte globulin; CsA, cyclosporin A; D, donor CMV serostatus; GCV, ganciclovir; R, recipient CMV serostatus; rATG, rabbit ATG.

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


Bacterial infection. No increase in the overall rate of bac- 114, 116121]. Infection occurred earlier among patients who
terial infection was noted with IL-2R antagonists, compared received alemtuzumab for graft-rejection therapy (72.5 days;
with other induction therapies or placebo (table 1) [17, 28, range, 2325 days) than among patients who received alem-
102104]. Higher mortality associated with infection was ob- tuzumab for induction therapy (120 days; range, 31328 days;
served among patients receiving daclizumab in a placebo-con- P p .09) [46]. Disseminated and pulmonary nocardiosis and
trolled study of cardiac transplant recipients who also received tuberculous and nontuberculous pneumonia (due to Myco-
cytolytic therapy (i.e., OKT3 and ATG) [30]. Infections in-
bacterium kansasii) have been described after treatment with
cluded sepsis (6 cases), CMV infection (1 case), and crypto-
alemtuzumab, mainly in patients who received alemtuzumab
coccal meningitis (1 case); the role of daclizumab in these com-
for treatment of acute graft rejection.
plications remains unclear [30].
Viral infection. Of 547 organ transplant recipients treated
CMV. Patients receiving IL-2R antagonists experience rates
with alemtuzumab without antiviral prophylaxis, 56 (10%) de-
of CMV infection that are similar to those among patients
receiving placebo or other induction therapies (table 1) [30, veloped 62 opportunistic infections [46], including 16 (26%)
96, 101103, 105108]. Receipt of cytolytic antigraft-rejection CMV infections, 12 (19%) BKV infections, and 3 (5%) EBV
treatment with basiliximab was associated with higher rates of infections with PTLD [46]. CMV disease occurred within 3
CMV infection, compared with receipt of ATG in renal trans- months after graft-rejection therapy with alemtuzumab [46].
plant recipients (17.5% vs. 7.8%; P p .02) [21]. Many patients (65%) who received alemtuzumab for treatment
EBV and EBV-induced PTLD. Most studies have not dem- of acute graft rejection also received another type of lympho-
onstrated an increased risk of PTLD after induction therapy cyte-depleting antibody, most commonly rATG [46]. In a sin-
with IL-2R antagonists (table 3) [17, 28, 57, 6870, 72, 108]. gle-center, 5-year trial of induction therapy with alemtuzumab
HCV. The effect of IL-2R antagonists on HCV infection is in renal transplantation, compared with other induction agents,

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


unknown. One study demonstrated higher rates of HCV in- a lower incidence of CMV infection was demonstrated with
fection recurrence among liver recipients after receipt of da- alemtuzumab using acyclovir for CMV prophylaxis in high-
clizumab [109], whereas other studies have demonstrated no risk recipients [74]. No tissue-invasive CMV infection, PTLD,
impact [110112]. or BKV-associated nephropathy was demonstrated in pediatric
Fungal infection. The incidence of fungal infection among
kidney recipients treated with alemtuzumab induction [122].
recipients of IL-2R antagonists was similar to the incidence
In a retrospective study of induction therapy in renal trans-
among recipients of placebo, no induction therapy, or other
plantation comparing alemtuzumab with ATGs (rabbit and
induction therapies (table 1) [21, 22, 26, 30, 31, 96, 102, 103,
equine) and IL-2R antagonists, alemtuzumab therapy was as-
105, 107, 113, 114].
sociated with a greater incidence of CMV infection, compared
with that in the IL-2R antagonist group (24.6% vs. 9.3%;
ALEMTUZUMAB
P p .01), with a rate similar to that in the rATG group [114].
Alemtuzumab is a humanized monoclonal antibody directed There was also a trend toward more BKV infections among
against the membrane glycoprotein CD52 on lymphocytes (T patients receiving alemtuzumab [114].
and B cells), monocytes and macrophages, and natural killer
Fungal infection. Invasive fungal infections have been de-
cells. Alemtuzumab achieves panT cell depletion; CD4 and
scribed after alemtuzumab therapy, including invasive asper-
CD8 cell counts reach a nadir 4 weeks after initiation of therapy
gillosis, mucormycosis, Scedosporium infection, Histoplasma
and remain at !25% of baseline values for 9 months [115].
capsulatum infection, Blastomyces dermatitidis infection, and
Data regarding infectious complications remain limited, be-
cryptococcal infection [46, 117, 123]. The risk of fungal infec-
cause alemtuzumab is used in induction therapy in a small
tion among transplant recipients treated with alemtuzumab
number of clinical centers. Some late invasive viral and fungal
infections have been observed in patients after induction and induction therapy appears to be similar to that for patients
graft-rejection therapy with alemtuzumab. Additional prospec- treated with other induction therapy agents [18, 114]. However,
tive data are needed. a greater risk of invasive fungal infection (mostly esophageal
Bacterial infection. Alemtuzumab is used at a lower dosage candidiasis) exists after treatment for acute graft rejection; most
in induction therapy for solid organ transplantation (2030 mg infections occurred within 3 months after the start of therapy
for 1 or 2 doses), compared with the dosage used for cancer [46].
therapy. Induction therapy with alemtuzumab is associated with Parasitic infection. Amebic meningitis due to Balamuthia
a low incidence of bacterial infection, compared with historical mandrillaris and Toxoplasma pneumonia has been described in
regimens or with other therapies (table 1) [18, 31, 34, 73, 74, patients receiving alemtuzumab for acute graft rejection [46].

IMMUNOCOMPROMISED HOSTS CID 2009:48 (15 March) 779


Table 3. Induction therapy and risk of posttransplantation lymphoproliferative disorder (PTLD).

Type of Duration of
Drug, study number Study type Database Period transplant follow-up RR (95% CI) of PTLD P Reference
hATG: 1 Retrospective data analysis SRTR (3752/41,686 recipients) 19962002 Kidney Censored 1.50 (0.932.43) .10 [67]
rATG
1 Retrospective data analysis SRTR (2376/41,686 recipients) 19962002 Kidney Censored 3.0 (1.535.89) .001 [67]
2 Randomized prospective study 90 patients 20022004 Kidney 15 months No PTLD [72]
3 Retrospective data analysis UNOS-OPTN (12,051/84,368 19872003 Kidney Median, 368 days 1.17 (0.871.58) .29 [71]
recipients)
4 Retrospective data analysis UNOS-OPTN (685/5072 19872003 Pediatric kidney Median, 368 days 1.51 (0.782.93) [71]
recipients)
5 Retrospective data analysis UNOS-OPTN (13,110/59,560 20002004 Kidney 730 days 1.63 .01 [69]

780
recipients)
L-IG
1 Retrospective data analysis UNOS-OPTN (8076/84,368 19872003 Kidney Median, 1433 days 1.61 (1.242.10) !.001 [71]
recipients)
2 Retrospective data analysis UNOS-OPTN (620/5072 19872003 Pediatric kidney Median, 1433 days 2.162 (1.223.82) .008 [71]
recipients)
ALG
1 Retrospective data analysis UNOS-OPTN (16,108/84,368 19872003 Kidney Median, 2055 days 1.35 (1.091.68) .006 [71]
recipients)
2 Retrospective data analysis UNOS-OPTN (1334/5072 19872003 Pediatric kidney Median, 2055 days 1.008 (0.581.76) [71]
recipients)
ATG (all)
1 Retrospective data analysis UNOS-OPTN (38,519 19972000 Kidney 727 days 1.29 (0.822.03) .27 [70]
recipients)
2 Retrospective data analysis USRDS (4353/25127 19962000 Kidney Censored 1.55 (1.2- 1.99) .001 [68]
recipients)
Basiliximab
1 Retrospective data analysis SRTR (5169/41,686 recipients) 19962002 Kidney Censored 1.83 (1.053.18) .032 [67]
2 Retrospective data analysis UNOS-OPTN (14,182/59,560 20002004 Kidney 730 days 0.84 .33 [69]
recipients)
Daclizumab

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


1 Retrospective data analysis SRTR (3377/41,686 recipients) 19962002 Kidney Censored 1.92 (1.083.41) .027 [67]
2 Randomized prospective study 90 patients 20022004 Kidney 15 months No PTLD [72]
3 Retrospective data analysis UNOS-OPTN (7511/59,560 20002004 Kidney 730 days 0.64 .06 [69]
recipients)
Anti-IL-2R as a
group
1 Retrospective data analysis UNOS-OPTN (7800/38,519 19972000 Kidney 727 days 1.14 (0.771.70) .52 [70]
recipients)
2 Retrospective data analysis USRDS (3936/25,127 19962000 Kidney Censored 1.16 (0.821.65) .39 [68]
recipients)
Alemtuzumab
1 Retrospective single-center 126 patients 19972003 Kidney Censored No PTLD [73]
study
2 Prospective single-center study 33 patients 19971998 Kidney 5 years 1 case [74]
3 Prospective single-center study 10 patients 20022003 Lung 6 months No PTLD [34]
4 Randomized prospective study 90 patients 20022004 Kidney 15 months No PTLD [72]
5 Retrospective data analysis UNOS-OPTN (1691/59,560) 20002004 Kidney 730 days 1.15 .74 [69]
recipients
Belatacept and bas- Randomized prospective study 218 patients 20012003 Kidney 13 months 3 casesa [75]
iliximab: 1

NOTE. ALG, antilymphocyte globulin; ATG, antithymocyte globulin; hATG, horse ATG; L-IG, lymphocyte immunoglobulin; rATG, rabbit ATG; RR, relative risk; SRTR, Scientific Registry of Transplant
Recipients; UNOS-OPTN, United Network for Organ Sharing and Organ Procurement and Transplant Network; USRDS, United States Renal Data System.

781
a
One patient who developed PTLD after receipt of belatacept also received OKT3 for treatment of acute graft rejection.

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


CHRONIC INDUCTION THERAPY AGENTS should be monitored by quantitative CMV load or antigenemia
(ABATACEPT AND BELATACEPT) assay every 2 weeks for 6 months after induction therapy in
the absence of anti-CMV prophylaxis. CMV monitoring and
Abatacept and belatacept are fusion proteins composed of Fc
prophylaxis (3 months) should be extended or reinstituted
fragments of a human IgG1 immunoglobulin linked to the
for at-risk individuals after the use of T celldepleting agents
extracellular domain of cytotoxic T lymphocyte antigen 4
for treatment of graft rejection (tables 1 and 2). For trans-
(CTLA-4) that block T cell activation by preventing T cell
plantations in which the donor and recipient are both CMV
costimulation. Experience with these agents remains limited.
seronegative, antiviral prophylaxis (e.g., famciclovir, valacyclo-
Three cases of PTLD have been reported in kidney transplant
vir, or acyclovir) for 46 months is recommended for preven-
recipients who received intensive treatment with belatacept (ta-
tion of infection with herpes simplex virus or varicella-zoster
ble 3) [75].
virus.
EBV and EBV-induced PTLD. The association between T
RECOMMENDATIONS: MONITORING FOR cell depletion or chronic induction therapy (abatacept and be-
INFECTION AND PREVENTATIVE THERAPIES latacept) and a possible risk of PTLD supports the monitoring
Experience with antilymphocyte therapies has provided a pre- of EBV-seronegative recipients of organs from seropositive do-
dictability of infectious complications based on the intensity nors with use of quantitative molecular assays (plasma or whole
and duration of T cell depletion. Strategies should be developed blood) monthly for at least 1 year after induction therapy [124].
to monitor and prevent common infections associated with For patients with detectable EBV loads, consideration should
antilymphocyte therapies [124]. be given to decreasing the intensity of immunosuppression and
Bacterial and fungal infections. Patients receiving induc- to increasing the monitoring of EBV loads. Quantitative EBV
tion therapies do not require routine laboratory monitoring assays of whole blood have higher sensitivity than do serum

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


for bacterial or fungal infections. Because of the duration of assays; PTLD can develop in patients with low EBV loads [125].
effect of prophylaxis for pneumonia due to Pneumocystis carinii, BKV. Kidney recipients who are receiving induction ther-
it should be prolonged (duration, 612 months) when T cell apy with T celldepleting agents should be monitored for BKV
depleting agents are used in induction or graft rejection therapy. infection with use of a sensitive urine (either cytopathology for
Trimethoprim-sulfamethoxazole (single strength, 80/400 mg decoy cells or a quantitative, nucleic acidbased viral load assay)
daily) prevents pneumonia due to Pneumocystis species and or blood (DNA) screening test at least monthly for 6 months.
other opportunistic infections (i.e., infection due to Nocardia, Any unexplained increase in serum creatinine level should
prompt examination of a renal biopsy specimen with use of
Toxoplasma, or Listeria species). Other prophylactic regimens
immunostaining or in situ hybridization for the detection of
against pneumonia due to P. carinii do not offer the same
BKV [76, 81, 124, 126].
protection. Patients with active CMV or HCV infection who
HCV. In patients who are HCV seropositive, a quantitative
have poor allograft function or who are treated for graft re-
HCV RNA load measurement is recommended at baseline and
jection should receive prophylaxis for longer periods (i.e., 6
at 3, 6, 9, and 12 months after transplantation [124]. Quan-
months to life).
titative HCV load does not correlate with the degree of hepatic
Individuals receiving induction therapy with T celldepleting
injury [127], and examination of biopsy specimens will be
agents who have a history of exposure (i.e., seropositivity) to
needed to guide decisions regarding antiviral therapy [124].
endemic fungi (i.e., the fungi that cause histoplasmosis, coc-
cidioidosis, blastomycosis) or to Cryptococcus species should
SUMMARY
receive prophylaxis (duration, 612 months) with fluconazole
(200400 mg daily; for infection due to Coccidioides, Histo- Infectious risks are greater when antilymphocyte therapies are
plasma, or Cryptococcus species) or itraconazole (200 mg orally used in the treatment of graft rejection, compared with the use
daily; for infection due to Histoplasma or Blastomyces species), of these therapies in induction immunosuppression. Antiviral
with adjustment in dosages of any calcineurin inhibitors or prophylaxis will reduce the risk for CMV infection associated
rapamycin. Monitoring with use of antigen assays or immu- with induction therapy with ATG. BKV replication and BKV-
noassays for Histoplasma and Cryptococcus species (and more associated nephropathy occur earlier and are more common
recently for species that cause coccidioides and blastomycosis) with induction therapy. In contrast, patients who receive IL-
may be useful; cross-reactivity is common. Comprehensive 2R antagonists for induction therapy experience a lower inci-
long-term data on infections associated with IL-2R antagonists dence of infection overall and of PTLD. The use of T cell
are lacking. depleting agents (e.g., ATG, OKT3, and alemtuzumab) for
CMV prevention. Patients who are at risk of CMV infection graft-rejection or induction therapy should be linked to ap-
(i.e., either the organ donor or recipient is CMV seropositive) propriate monitoring and/or prophylaxis for pneumonia due

782 CID 2009:48 (15 March) IMMUNOCOMPROMISED HOSTS


to P. carinii and for CMV and fungal infection. Monitoring of 15. Waller EK, Langston AA, Lonial S, et al. Pharmacokinetics and phar-
macodynamics of anti-thymocyte globulin in recipients of partially
EBV in seronegative recipients of organs from EBV-seropositive HLA-matched blood hematopoietic progenitor cell transplantation.
donors and monitoring of BKV in renal transplant recipients Biol Blood Marrow Transplant 2003; 9:46071.
should be performed using quantitative assays for patients who 16. Alangaden GJ, Thyagarajan R, Gruber SA, et al. Infectious compli-
cations after kidney transplantation: current epidemiology and as-
are receiving T celldepleting agents.
sociated risk factors. Clin Transplant 2006; 20:4019.
17. Lebranchu Y, Bridoux F, Buchler M, et al. Immunoprophylaxis with
Acknowledgments basiliximab compared with antithymocyte globulin in renal transplant
patients receiving MMF-containing triple therapy. Am J Transplant
Potential conflicts of interest. J.A.F. has served as a consultant for 2002; 2:4856.
Bristol-Myers Squibb, Merck, Hoffman-La Roche, T2 Biosystems, Astellas, 18. Ashcraft E, Farney A, Moore P, et al. A prospective randomized single
and FDA/CBERXenotransplantation; has served on the scientific advisory center trial of alemtuzumab versus rabbit anti-thymocyte induction
board for Primera; has served on the speakers bureau for Hoffman-La in renal and pancreas transplantation: infectious complications [ab-
Roche; and has received corporate grant support from PATH Alliance (Ax- stract 335]. In: Program and abstracts of the 7th American Transplant
iom/Astellas). J.A.F. has patents for molecular cloning of antigens shared Congress (San Francisco). 2007:234.
by rat- and human-derived Pneumocystis carinii (patent 5442050; GenBank 19. Schwartz JJ, Ishitani MB, Weckwerth J, Morgenstern B, Milliner D,
accession numbers L43850, L43851, and L43852) and molecular sequence Stegall MD. Decreased incidence of acute rejection in adolescent kid-
of swine retrovirus and methods of use (patent 6190861; patent cooperation ney transplant recipients using antithymocyte induction and triple
treaty international patent WO07/21836; GenBank accession numbers immunosuppression. Transplantation 2007; 84:71521.
AF038600, AF038601, and AF038599). N.C.I.: no conflicts. 20. Abou-Jaoude MM, Ghantous I, Najm R, Afif C, Almawi WY. Dacli-
zumab versus anti-thymocyte globulin-fresenius as induction therapy
for low-risk kidney transplant recipients. Transplant Proc 2003; 35:
References
27312.
1. Parasuraman R, Nizaar A, Karthikeyan V, et al. Significant increase 21. Brennan DC, Daller JA, Lake KD, Cibrik D, Del Castillo D. Rabbit
in infection-associated renal allograft failure over the past decade: antithymocyte globulin versus basiliximab in renal transplantation. N
UNOS data analysis [abstract 146]. In: Program and abstracts of the Engl J Med 2006; 355:196777.

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


8th American Transplant Congress (Toronto, Ontario, Canada). 2008: 22. Carlsen J, Johansen M, Boesgaard S, et al. Induction therapy after
218. cardiac transplantation: a comparison of anti-thymocyte globulin and
2. Ciancio G, Burke GW 3rd. Alemtuzumab (Campath-1H) in kidney daclizumab in the prevention of acute rejection. J Heart Lung Trans-
transplantation. Am J Transplant 2008; 8:1520. plant 2005; 24:296302.
3. Kaden J. Eleven years intraoperative ATG bolus: a list of successes. 23. Mariat C, Afiani A, Alamartine E, Thibaudin D, de Filippis JP, Ber-
Ann Transplant 2002; 7:410. thoux F. A pilot study comparing basiliximab and anti-thymocyte
4. Feng X, Kajigaya S, Solomou EE, et al. Rabbit ATG but not horse globulin as induction therapy in sensitized renal allograft recipients.
ATG promotes expansion of functional CD4+CD25highFOXP3+ reg- Transplant Proc 2001; 33:31923.
ulatory T cells in vitro. Blood 2008; 111:367583. 24. Mourad G, Garrigue V, Squifflet JP, et al. Induction versus nonin-
5. Raefsky EL, Gascon P, Gratwohl A, Speck B, Young NS. Biological duction in renal transplant recipients with tacrolimus-based immu-
and immunological characterization of ATG and ALG. Blood 1986; nosuppression. Transplantation 2001; 72:10505.
68:7129. 25. Hartwig MG, Snyder LD, Appel JZ III, et al. Rabbit anti-thymocyte
6. Bourdage JS, Hamlin DM. Comparative polyclonal antithymocyte globulin induction therapy does not prolong survival after lung trans-
globulin and antilymphocyte/antilymphoblast globulin anti-CD an- plantation. J Heart Lung Transplant 2008; 27:54753.
tigen analysis by flow cytometry. Transplantation 1995; 59:1194200. 26. Mattei MF, Redonnet M, Gandjbakhch I, et al. Lower risk of infectious
7. Hardinger KL, Schnitzler MA, Miller B, et al. Five-year follow up of deaths in cardiac transplant patients receiving basiliximab versus anti-
thymoglobulin versus ATGAM induction in adult renal transplan- thymocyte globulin as induction therapy. J Heart Lung Transplant
tation. Transplantation 2004; 78:13641. 2007; 26:6939.
8. Agha IA, Rueda J, Alvarez A, et al. Short course induction immu- 27. Abou-Ayache R, Buchler M, Lepogamp P, et al. CMV infections after
nosuppression with thymoglobulin for renal transplant recipients. two doses of daclizumab versus thymoglobulin in renal transplant
Transplantation 2002; 73:4735. patients receiving mycophenolate mofetil, steroids and delayed cy-
9. Brennan DC, Flavin K, Lowell JA, et al. A randomized, double-blinded closporine A. Nephrol Dial Transplant 2008; 23:202432.
comparison of thymoglobulin versus atgam for induction immuno- 28. Sollinger H, Kaplan B, Pescovitz MD, et al. Basiliximab versus an-
suppressive therapy in adult renal transplant recipients. Transplan- tithymocyte globulin for prevention of acute renal allograft rejection.
tation 1999; 67:10118. Transplantation 2001; 72:19159.
10. Peddi VR, Bryant M, Roy-Chaudhury P, Woodle ES, First MR. Safety, 29. Mullen JC, Oreopoulos A, Lien DC, et al. A randomized, controlled
efficacy, and cost analysis of thymoglobulin induction therapy with trial of daclizumab vs. anti-thymocyte globulin induction for lung
intermittent dosing based on CD3+ lymphocyte counts in kidney and transplantation. J Heart Lung Transplant 2007; 26:50410.
kidney-pancreas transplant recipients. Transplantation 2002; 73: 30. Hershberger RE, Starling RC, Eisen HJ, et al. Daclizumab to prevent
15148. rejection after cardiac transplantation. N Engl J Med 2005; 352:
11. Starzl TE, Murase N, Abu-Elmagd K, et al. Tolerogenic immunosup- 270513.
pression for organ transplantation. Lancet 2003; 361:150210. 31. Ciancio G, Sageshima J, Burke GW. Evaluation of a randomized trial
12. Bunn D, Lea CK, Bevan DJ, Higgins RM, Hendry BM. The phar- of three induction antibodies in deceased donor renal transplantation
macokinetics of anti-thymocyte globulin (ATG) following intravenous at 18 months follow-up [abstract 919]. In: Program and abstracts of
infusion in man. Clin Nephrol 1996; 45:2932. the 1st World Transplant Congress (Boston). 2006:376.
13. Mohty M. Mechanisms of action of antithymocyte globulin: T-cell 32. Clark G, Walsh G, Deshpande P, Koffman G. Improved efficacy of
depletion and beyond. Leukemia 2007; 21:138794. basiliximab over antilymphocyte globulin induction therapy in pae-
14. Louis S, Audrain M, Cantarovich D, et al. Long-term cell monitoring diatric renal transplantation. Nephrol Dial Transplant 2002; 17:
of kidney recipients after an antilymphocyte globulin induction with 13049.
and without steroids. Transplantation 2007; 83:71221. 33. Palmer SM, Miralles AP, Lawrence CM, Gaynor JW, Davis RD, Tapson

IMMUNOCOMPROMISED HOSTS CID 2009:48 (15 March) 783


VF. Rabbit antithymocyte globulin decreases acute rejection after lung proliferative disorder localized near the allograft in renal transplan-
transplantation: results of a randomized, prospective study. Chest tation. Transplantation 2000; 69:80914.
1999; 116:12733. 55. Younes BS, McDiarmid SV, Martin MG, et al. The effect of immu-
34. McCurry KR, Iacono A, Zeevi A, et al. Early outcomes in human nosuppression on posttransplant lymphoproliferative disease in pe-
lung transplantation with thymoglobulin or Campath-1H for recip- diatric liver transplant patients. Transplantation 2000; 70:949.
ient pretreatment followed by posttransplant tacrolimus near-mono- 56. Winter P, Schoeneich G, Miersch WD, Klehr HU. Tumour induction
therapy. J Thorac Cardiovasc Surg 2005; 130:52837. as a consequence of immunosuppression after renal transplantation.
35. Pourmand G, Salem S, Mehrsai A, Taherimahmoudi M, Ebrahimi R, Int Urol Nephrol 1997; 29:7019.
Pourmand MR. Infectious complications after kidney transplantation: 57. Opelz G, Dohler B. Lymphomas after solid organ transplantation: a
a single-center experience. Transpl Infect Dis 2007; 9:3029. collaborative transplant study report. Am J Transplant 2004; 4:22230.
36. Shoja MM, Ardalan MR, Etemadi J, Tubbs RS, Varshochi M. Renal 58. Webber SA, Naftel DC, Fricker FJ, et al. Lymphoproliferative disorders
allograft abscesses following transplant: case report and literature re- after paediatric heart transplantation: a multi-institutional study. Lan-
view. Exp Clin Transplant 2007; 5:7203. cet 2006; 367:2339.
37. Bijleveld CG, Klompmaker IJ, van den Berg AP, et al. Incidence, risk 59. Walker RC, Marshall WF, Strickler JG, et al. Pretransplantation as-
factors, and outcome of antithymocyte globulin treatment of steroid- sessment of the risk of lymphoproliferative disorder. Clin Infect Dis
resistant rejection after liver transplantation. Transpl Int 1996; 9: 1995; 20:134653.
5705. 60. Walker RC, Paya CV, Marshall WF, et al. Pretransplantation sero-
38. Jamil B, Nicholls KM, Becker GJ, Walker RG. Influence of anti-re- negative Epstein-Barr virus status is the primary risk factor for post-
jection therapy on the timing of cytomegalovirus disease and other transplantation lymphoproliferative disorder in adult heart, lung, and
infections in renal transplant recipients. Clin Transplant 2000; 14: other solid organ transplantations. J Heart Lung Transplant 1995; 14:
148. 21421.
39. Zamora MR. Controversies in lung transplantation: management of 61. Patel H, Vogl DT, Aqui N, et al. Posttransplant lymphoproliferative
cytomegalovirus infections. J Heart Lung Transplant 2002; 21:8419. disorder in adult liver transplant recipients: a report of seventeen
40. Huurman VA, Kalpoe JS, van de Linde P, et al. Choice of antibody cases. Leuk Lymphoma 2007; 48:88591.
immunotherapy influences cytomegalovirus viremia in simultaneous 62. Armitage JM, Kormos RL, Stuart RS, et al. Posttransplant lympho-
pancreas-kidney transplant recipients. Diabetes Care 2006; 29:8427. proliferative disease in thoracic organ transplant patients: ten years
41. Ozaki KS, Pestana JO, Granato CF, Pacheco-Silva A, Camargo LF. of cyclosporine-based immunosuppression. J Heart Lung Transplant

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


Sequential cytomegalovirus antigenemia monitoring in kidney trans- 1991; 10:87786; discussion, 867.
plant patients treated with antilymphocyte antibodies. Transpl Infect 63. Holmes RD, Sokol RJ. Epstein-Barr virus and post-transplant lym-
Dis 2004; 6:638. phoproliferative disease. Pediatr Transplant 2002; 6:45664.
42. Buchler M, Hurault de Ligny B, Madec C, Lebranchu Y. Induction 64. Preiksaitis JK. New developments in the diagnosis and management
therapy by anti-thymocyte globulin (rabbit) in renal transplantation: of posttransplantation lymphoproliferative disorders in solid organ
a 1-yr follow-up of safety and efficacy. Clin Transplant 2003; 17: transplant recipients. Clin Infect Dis 2004; 39:101623.
53945. 65. Paya CV, Fung JJ, Nalesnik MA, et al. Epstein-Barr virusinduced
43. Docke WD, Prosch S, Fietze E, et al. Cytomegalovirus reactivation posttransplant lymphoproliferative disorders. ASTS/ASTP EBV-PTLD
and tumour necrosis factor. Lancet 1994; 343:2689. Task Force and the Mayo Clinic Organized International Consensus
44. Fietze E, Prosch S, Reinke P, et al. Cytomegalovirus infection in trans- Development Meeting. Transplantation 1999; 68:151725.
plant recipients: the role of tumor necrosis factor. Transplantation 66. Gaber AO, First MR, Tesi RJ, et al. Results of the double-blind, ran-
1994; 58:67580. domized, multicenter, phase III clinical trial of thymoglobulin versus
45. Kaden J, Eichler S, May G, Strobelt V. The ATG bolus does not induce Atgam in the treatment of acute graft rejection episodes after renal
a persistent inversion of the CD4/8 ratio. Transplant Proc 2002; 34: transplantation. Transplantation 1998; 66:2937.
237981. 67. Bustami RT, Ojo AO, Wolfe RA, et al. Immunosuppression and the
46. Peleg AY, Husain S, Kwak EJ, et al. Opportunistic infections in 547 risk of post-transplant malignancy among cadaveric first kidney trans-
organ transplant recipients receiving alemtuzumab, a humanized plant recipients. Am J Transplant 2004; 4:8793.
monoclonal CD-52 antibody. Clin Infect Dis 2007; 44:20412. 68. Caillard S, Dharnidharka V, Agodoa L, Bohen E, Abbott K. Post-
47. von Muller L, Schliep C, Storck M, et al. Severe graft rejection, in- transplant lymphoproliferative disorders after renal transplantation in
creased immunosuppression, and active CMV infection in renal trans- the United States in era of modern immunosuppression. Transplan-
plantation. J Med Virol 2006; 78:3949. tation 2005; 80:123343.
48. Mihalov ML, Gattuso P, Abraham K, Holmes EW, Reddy V. Incidence 69. Cherikh W, Ring M, Kauffman HM, et al. Updated analysis of dis-
of post-transplant malignancy among 674 solid-organtransplant re- sociation of depletion and PTLD in kidney recipients treated with
cipients at a single center. Clin Transplant 1996; 10:24855. alemtuzumab induction therapy [abstract 332]. In: Program and ab-
49. Penn I. Posttransplantation de novo tumors in liver allograft recip- stracts of the 7th American Transplant Congress (San Francisco).
ients. Liver Transpl Surg 1996; 2:529. 2007:233.
50. Penn I. De novo malignances in pediatric organ transplant recipients. 70. Cherikh WS, Kauffman HM, McBride MA, Maghirang J, Swinnen
Pediatr Transplant 1998; 2:5663. LJ, Hanto DW. Association of the type of induction immunosup-
51. Jain AB, Yee LD, Nalesnik MA, et al. Comparative incidence of de pression with posttransplant lymphoproliferative disorder, graft sur-
novo nonlymphoid malignancies after liver transplantation under tac- vival, and patient survival after primary kidney transplantation. Trans-
rolimus using surveillance epidemiologic end result data. Transplan- plantation 2003; 76:128993.
tation 1998; 66:1193200. 71. Dharnidharka VR, Stevens G. Risk for post-transplant lymphopro-
52. Ondrus D, Pribylincova V, Breza J, et al. The incidence of tumours liferative disorder after polyclonal antibody induction in kidney trans-
in renal transplant recipients with long-term immunosuppressive plantation. Pediatr Transplant 2005; 9:6226.
therapy. Int Urol Nephrol 1999; 31:41722. 72. Ciancio G, Burke GW, Gaynor JJ, et al. A randomized trial of three
53. Winkelhorst JT, Brokelman WJ, Tiggeler RG, Wobbes T. Incidence renal transplant induction antibodies: early comparison of tacrolimus,
and clinical course of de-novo malignancies in renal allograft recip- mycophenolate mofetil, and steroid dosing, and newer immune-mon-
ients. Eur J Surg Oncol 2001; 27:40913. itoring. Transplantation 2005; 80:45765.
54. Kew CE 2nd, Lopez-Ben R, Smith JK, et al. Postransplant lympho- 73. Knechtle SJ, Fernandez LA, Pirsch JD, et al. Campath-1H in renal

784 CID 2009:48 (15 March) IMMUNOCOMPROMISED HOSTS


transplantation: the University of Wisconsin experience. Surgery dence of lymphoproliferative disorder after immunosuppression with
2004; 136:75460. the monoclonal antibody OKT3 in cardiac-transplant recipients. N
74. Watson CJ, Bradley JA, Friend PJ, et al. Alemtuzumab (CAMPATH Engl J Med 1990; 323:17238.
1H) induction therapy in cadaveric kidney transplantationefficacy 94. Jensen PB, Birkeland SA, Rohrp N, Elbirk A, Jorgensen KA. Devel-
and safety at five years. Am J Transplant 2005; 5:134753. opment of anti-OKT3 antibodies after OKT3 treatment. Scand J Urol
75. Vincenti F, Larsen C, Durrbach A, et al. Costimulation blockade with Nephrol 1996; 30:22730.
belatacept in renal transplantation. N Engl J Med 2005; 353:77081. 95. Nashan B. Antibody induction therapy in renal transplant patients
76. Hirsch HH, Brennan DC, Drachenberg CB, et al. Polyomavirus-as- receiving calcineurin-inhibitor immunosuppressive regimens: a com-
sociated nephropathy in renal transplantation: interdisciplinary anal- parative review. BioDrugs 2005; 19:3946.
yses and recommendations. Transplantation 2005; 79:127786. 96. Nashan B, Moore R, Amlot P, Schmidt AG, Abeywickrama K, Sou-
77. Dadhania D, Snopkowski C, Ding R, et al. Epidemiology of BK virus lillou JP. Randomised trial of basiliximab versus placebo for control
in renal allograft recipients: independent risk factors for BK virus of acute cellular rejection in renal allograft recipients. CHIB 201 In-
replication. Transplantation 2008; 86:5218. ternational Study Group. Lancet 1997; 350:11938.
78. Dharnidharka V, Cherikh W. Analysis of OPTN/UNOS data for rate 97. Amlot PL, Rawlings E, Fernando ON, et al. Prolonged action of a
of treatment for, and risk factors for, BK virus nephropathy (BKVN) chimeric interleukin-2 receptor (CD25) monoclonal antibody used
in kidney transplant recipients [abstract 390]. In: Program and ab- in cadaveric renal transplantation. Transplantation 1995; 60:74856.
stracts of the 8th American Transplant Congress (Toronto, Ontario, 98. Kovarik J, Breidenbach T, Gerbeau C, Korn A, Schmidt AG, Nashan
Canada). 2008:283. B. Disposition and immunodynamics of basiliximab in liver allograft
79. Smith JM, Dharnidharka VR, Talley L, Martz K, McDonald RA. BK recipients. Clin Pharmacol Ther 1998; 64:6672.
virus nephropathy in pediatric renal transplant recipients: an analysis 99. Kovarik JM, Rawlings E, Sweny P, et al. Prolonged immunosuppressive
of the North American Pediatric Renal Trials and Collaborative Stud- effect and minimal immunogenicity from chimeric (CD25) mono-
ies (NAPRTCS) registry. Clin J Am Soc Nephrol 2007; 2:103742. clonal antibody SDZ CHI 621 in renal transplantation. Transplant
80. Srinivas TR, Schold JD, Eagan GA, et al. Total rabbit ATG dose is a Proc 1996; 28:9134.
risk factor for onset of BK viremia in Kidney transplant recipients 100. Sterkers G, Baudouin V, Ansart-Pirenne H, et al. Duration of action
[abstract 336]. In: Program and abstracts of the 7th American Trans- of a chimeric interleukin-2 receptor monoclonal antibody, basilixi-
plant Congress (San Francisco). 2007:234. mab, in pediatric kidney transplant recipients. Transplant Proc
81. Hirsch HH, Knowles W, Dickenmann M, et al. Prospective study of 2000; 32:27579.

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


polyomavirus type BK replication and nephropathy in renal-trans- 101. Vincenti F. Potential of daclizumab in solid organ transplantation.
plant recipients. N Engl J Med 2002; 347:48896. BioDrugs 1999; 11:33341.
82. Fabrizi F, Martin P, Dixit V, Bunnapradist S, Dulai G. Hepatitis C 102. Nashan B, Light S, Hardie IR, Lin A, Johnson JR. Reduction of acute
virus antibody status and survival after renal transplantation: meta- renal allograft rejection by daclizumab. Daclizumab Double Therapy
analysis of observational studies. Am J Transplant 2005; 5:145261. Study Group. Transplantation 1999; 67:1105.
83. Meier-Kriesche HU, Ojo AO, Hanson JA, Kaplan B. Hepatitis C an- 103. Vincenti F, Kirkman R, Light S, et al. Interleukin-2receptor blockade
tibody status and outcomes in renal transplant recipients. Transplan- with daclizumab to prevent acute rejection in renal transplantation.
tation 2001; 72:2414. Daclizumab Triple Therapy Study Group. N Engl J Med 1998; 338:
84. Luan FL, Schaubel DE, Zhang H, et al. Impact of immunosuppressive 1615.
regimen on survival of kidney transplant recipients with hepatitis C. 104. Mehra MR, Zucker MJ, Wagoner L, et al. A multicenter, prospective,
Transplantation 2008; 85:16016. randomized, double-blind trial of basiliximab in heart transplanta-
85. Sener A, Barth RN, Cooper M, et al. T-cell depletion does not po- tion. J Heart Lung Transplant 2005; 24:1297304.
tentiate the progression of liver cirrhosis in HCV+ renal transplant 105. Ponticelli C, Yussim A, Cambi V, et al. A randomized, double-blind
patients [abstract 140]. In: Program and abstracts of the 8th American trial of basiliximab immunoprophylaxis plus triple therapy in kidney
Transplant Congress (Toronto, Ontario, Canada). 2008:216. transplant recipients. Transplantation 2001; 72:12617.
86. Wheat LJ, Connolly P, Durkin M, et al. False-positive Histoplasma 106. Lawen JG, Davies EA, Mourad G, et al. Randomized double-blind
antigenemia caused by antithymocyte globulin antibodies. Transpl study of immunoprophylaxis with basiliximab, a chimeric anti
Infect Dis 2004; 6:237. interleukin-2 receptor monoclonal antibody, in combination with
87. Wheat LJ, Connolly P, Durkin M, Book BK, Pescovitz MD. Elimi- mycophenolate mofetil-containing triple therapy in renal trans-
nation of false-positive Histoplasma antigenemia caused by human plantation. Transplantation 2003; 75:3743.
anti-rabbit antibodies in the second-generation Histoplasma antigen 107. Kahan BD, Rajagopalan PR, Hall M. Reduction of the occurrence of
assay. Transpl Infect Dis 2006; 8:21921. acute cellular rejection among renal allograft recipients treated with
88. Abbott KC, Hypolite I, Poropatich RK, et al. Hospitalizations for basiliximab, a chimeric antiinterleukin-2receptor monoclonal an-
fungal infections after renal transplantation in the United States. tibody. United States Simulect Renal Study Group. Transplantation
Transpl Infect Dis 2001; 3:20311. 1999; 67:27684.
89. Hibberd PL, Tolkoff-Rubin NE, Cosimi AB, et al. Symptomatic cy- 108. Webster AC, Playford EG, Higgins G, Chapman JR, Craig JC. Inter-
tomegalovirus disease in the cytomegalovirus antibody seropositive leukin 2 receptor antagonists for renal transplant recipients: a meta-
renal transplant recipient treated with OKT3. Transplantation 1992; analysis of randomized trials. Transplantation 2004; 77:16676.
53:6872. 109. Nelson DR, Soldevila-Pico C, Reed A, et al. Antiinterleukin-2 re-
90. Oh CS, Stratta RJ, Fox BC, Sollinger HW, Belzer FO, Maki DG. ceptor therapy in combination with mycophenolate mofetil is asso-
Increased infections associated with the use of OKT3 for treatment ciated with more severe hepatitis C recurrence after liver transplan-
of steroid-resistant rejection in renal transplantation. Transplantation tation. Liver Transpl 2001; 7:106470.
1988; 45:6873. 110. Kato T, Gaynor JJ, Yoshida H, et al. Randomized trial of steroid-free
91. Thistlethwaite JR Jr, Stuart JK, Mayes JT, et al. Complications and induction versus corticosteroid maintenance among orthotopic liver
monitoring of OKT3 therapy. Am J Kidney Dis 1988; 11:1129. transplant recipients with hepatitis C virus: impact on hepatic fibrosis
92. Meier-Kriesche HU, Arndorfer JA, Kaplan B. Association of antibody progression at one year. Transplantation 2007; 84:82935.
induction with short- and long-term cause-specific mortality in renal 111. Heffron TG, Smallwood GA, de Vera ME, Davis L, Martinez E, Stieber
transplant recipients. J Am Soc Nephrol 2002; 13:76972. AC. Daclizumab induction in liver transplant recipients. Transplant
93. Swinnen LJ, Costanzo-Nordin MR, Fisher SG, et al. Increased inci- Proc 2001; 33:1527.

IMMUNOCOMPROMISED HOSTS CID 2009:48 (15 March) 785


112. Filipponi F, Callea F, Salizzoni M, et al. Double-blind comparison of path-1H) for immunosuppressive therapy in organ transplantation.
hepatitis C histological recurrence rate in HCV+ liver transplant re- Transpl Int 2006; 19:70514.
cipients given basiliximab + steroids or basiliximab + placebo, in 120. Ortiz J, Palma-Vargas J, Wright F, et al. Campath induction for kidney
addition to cyclosporine and azathioprine. Transplantation 2004; 78: transplantation: report of 297 cases. Transplantation 2008; 85:15506.
148895. 121. Kaufman DB, Leventhal JR, Gallon LG, Parker MA. Alemtuzumab
113. Stratta RJ, Alloway RR, Lo A, Hodge EE. A prospective, randomized, induction and prednisone-free maintenance immunotherapy in si-
multicenter study evaluating the safety and efficacy of two dosing multaneous pancreas-kidney transplantation comparison with rabbit
regimens of daclizumab compared to no antibody induction in si- antithymocyte globulin inductionlong-term results. Am J Trans-
multaneous kidney-pancreas transplantation: results at 3 years. Trans- plant 2006; 6:3319.
plant Proc 2005; 37:35314. 122. Tan HP, Ellis D, Moritz ML, et al. Pediatric living donor kidney
114. Schadde E, DAlessandro AM, Knechtle SJ, et al. Alemtuzumab in- transplantation using alemtuzumab pre-conditioning and tacrolimus
duction and triple maintenance immunotherapy in kidney trans- monotherapy [abstract 55]. In: Program and abstracts of the 8th
plantation from donors after cardiac death. Transpl Int 2008; 21: American Transplant Congress (Toronto, Ontario, Canada). 2008:193.
62536. 123. Gauthier GM, Safdar N, Klein BS, Andes DR. Blastomycosis in solid
115. Morris EC, Rebello P, Thomson KJ, et al. Pharmacokinetics of alem- organ transplant recipients. Transpl Infect Dis 2007; 9:3107.
tuzumab used for in vivo and in vitro T-cell depletion in allogeneic 124. Humar A, Michaels M. American Society of Transplantation rec-
transplantations: relevance for early adoptive immunotherapy and ommendations for screening, monitoring and reporting of infectious
infectious complications. Blood 2003; 102:4046. complications in immunosuppression trials in recipients of organ
116. Tzakis AG, Kato T, Nishida S, et al. Alemtuzumab (Campath-1H) transplantation. Am J Transplant 2006; 6:26274.
combined with tacrolimus in intestinal and multivisceral transplan- 125. Kullberg-Lindh C, Olofsson S, Brune M, Lindh M. Comparison of
tation. Transplantation 2003; 75:15127. serum and whole blood levels of cytomegalovirus and Epstein-Barr
117. Nath DS, Kandaswamy R, Gruessner R, Sutherland DE, Dunn DL, virus DNA. Transpl Infect Dis 2008; 10:30815.
Humar A. Fungal infections in transplant recipients receiving alem- 126. Nickeleit V, Klimkait T, Binet IF, et al. Testing for polyomavirus type
tuzumab. Transplant Proc 2005; 37:9346. BK DNA in plasma to identify renal-allograft recipients with viral
118. Malek SK, Obmann MA, Gotoff RA, Foltzer MA, Hartle JE, Potdar nephropathy. N Engl J Med 2000; 342:130915.
S. Campath-1H induction and the incidence of infectious compli- 127. McCormick SE, Goodman ZD, Maydonovitch CL, Sjogren MH. Eval-
cations in adult renal transplantation. Transplantation 2006; 81:1720. uation of liver histology, ALT elevation, and HCV RNA titer in patients

Downloaded from cid.oxfordjournals.org by guest on May 11, 2011


119. Magliocca JF, Knechtle SJ. The evolving role of alemtuzumab (Cam- with chronic hepatitis C. Am J Gastroenterol 1996; 91:151622.

786 CID 2009:48 (15 March) IMMUNOCOMPROMISED HOSTS

Das könnte Ihnen auch gefallen