Sie sind auf Seite 1von 18

In-Depth Review

Medical Care of Kidney Transplant Recipients after the First


Posttransplant Year
Arjang Djamali,* Millie Samaniego,* Brenda Muth,* Rebecca Muehrer,*
R. Michael Hofmann,* John Pirsch,* Andrew Howard,

Georges Mourad,

and
Bryan N. Becker*
*Department of Medicine, Nephrology Section, University of Wisconsin, Madison, Madison, Wisconsin;

Metropolitan
Nephrology Associates, Alexandria, Virginia; and

Montpellier Medical School, Montpellier, France
Kidney transplantation is the treatment of choice for patients with ESRD. Despite improvements in short-term patient and
graft outcomes, there has been no major improvement in long-term outcomes. The use of kidney allografts from expanded-
criteria donors, polyoma virus nephropathy, underimmunosuppression, and incomplete functional recovery after rejection
episodes may play a role in the lack of improvement in long-term outcomes. Other factors, including cardiovascular disease,
infections, and malignancies, also shorten patient survival and therefore reduce the functional life of an allograft. There is a
need for interventions that improve long-term outcomes in kidney transplant recipients. These patients are a unique subset
of patients with chronic kidney disease. Therefore, interventions need to address disease progression, comorbid conditions,
and patient mortality through a multifaceted approach. The Kidney Disease Outcomes Quality Initiative from the National
Kidney Foundation, the European Best Practice Guidelines, and the forthcoming Kidney Disease: Improving Global Out-
comes clinical practice guidelines can serve as a cornerstone of this approach. The unique aspects of chronic kidney disease
in the transplant recipient require the integration of specific transplant-oriented problems into this care schema and a concrete
partnership among transplant centers, community nephrologists, and primary care physicians. This article reviews the
contemporary aspects of care for these patients.
Clin J Am Soc Nephrol 1: 623640, 2006. doi: 10.2215/CJN.01371005
W
ith tremendous advances in the fields of transplant
surgery, medicine, and pharmacology during the
past 50 yr, kidney transplantation has become the
treatment of choice for patients with ESRD (13). Currently, one
of every four patients with ESRD in the United States is a
kidney transplant recipient (4). A considerable part of this
success is due to better short-term outcomes that have resulted
from improved management of early postsurgical complica-
tions and prevention of acute rejection episodes (5). However,
new evidence demonstrates that there has been a lack of sig-
nificant improvement in long-term outcomes (2,57). Earlier
studies had reported projected half-lives of 13.8 yr for deceased-
donor transplants that were performed between 1988 and 1996
(8), yet, recent analyses covering the same interval demon-
strated a more modest actual graft half-life of 8 yr (6). These
data on long-term graft survival are even more startling when
one considers that deceased-donor transplants that were per-
formed between 1995 and 2000 manifest a trend toward worse
survival (5), that there is a direct association between chronic
allograft dysfunction and increased cardiovascular mortality
(9), and that graft loss has a detrimental impact on patient
survival (Figure 1) (1,10).
Why have long-term outcomes in kidney transplant recipi-
ents not improved? The use of kidney allografts with varying
quality, e.g., extended-criteria donors, polyoma/BK virus ne-
phropathy, underimmunosuppression, and incomplete recov-
ery of kidney function after rejection episodes in the first post-
transplant year has been invoked (5,6,11). However, other
factors, including cardiovascular disease (CVD), infections, and
malignancies, contribute to reduced patient survival and there-
fore a reduction in the functional life of an allograft (Tables 1
and 2) (9,10,1214). Even when these comorbid complications
lead only to illness, they may take a toll on allograft survival
(9,10,12). As a result, interventions that aim to improving long-
term outcomes in kidney transplant recipients should address
disease progression, comorbid conditions, and patient mortal-
ity. This multifaceted task may be accomplished best through a
partnership among transplant centers, community nephrolo-
gists, and primary care physicians, who already are or inevita-
bly will be involved in the care of kidney transplant recipients.
Kidney transplant recipients are a unique subset of patients
with chronic kidney disease (CKD) (1416). Most of these pa-
tient have stage 2 or 3 CKD, based on the National Kidney
Foundation (NKF) CKD classification schema even immedi-
ately posttransplantation (1518); a large percentage of patients
initiate their transplant function with an estimated GFR (eGFR)
that even may be as low as 30 ml/min (17). Although disease
Published online ahead of print. Publication date available at www.cjasn.org.
Address correspondence to: Dr. Arjang Djamali, University of Wisconsin Madi-
son, School of Medicine, 3034 Fish Hatchery Road, Suite B, Madison, WI 53713.
Phone: 608-270-5681; Fax: 608-270-5677; E-mail: axd@medicine.wisc.edu
Copyright 2006 by the American Society of Nephrology ISSN: 1555-9041/104-0623
progression rates (reported as the mean slope of creatinine
clearance or eGFR) have consistently been reported in the range
of 1.4 to 2.4 ml/min per yr (16,18,19), some patients lose
function more rapidly (5 to 20 ml/min per yr), whereas others
stabilize their GFR (18,19). The rate of loss of function does not
seem to be associated with baseline function of the transplant.
Finally, morbidity and mortality rates increase significantly
from stage 1 to stage 5 CKD in kidney transplant recipients
(15,16). At the same level of kidney function (stage 3), death
rates are higher in kidney transplant recipients compared with
nontransplant patients with CKD (16). Such data suggest that
despite a slower decline in kidney function over time, kidney
transplant recipients have marked differences in comorbidity
and mortality rates compared with their CKD counterparts. As
such, the impact of a 1-ml/min per yr decline in GFR has a
greater impact on long-term outcomes in kidney transplant
recipients.
Kidney transplant recipients return to a higher level of phys-
ical functioning compared with their counterparts on dialysis.
Given this, it is important to recognize that immunosuppres-
sion and graft function are only one component of their health
care. We propose that the objective should be a systematic
integration of transplant, CKD, and general health care. The
NKF Kidney Disease Outcomes Quality Initiative or K/DOQI
(http://www.kidney.org/professionals/kdoqi/guidelines.cfm),
the European Best Practice Guidelines (EBPG; http://www.
ndt-educational.org/guidelines.asp), and the forthcoming In-
ternational Kidney Disease Improving Global Outcomes
(KDIGO; http://www.kdigo.org/welcome.htm) clinical prac-
tice guidelines should be generalized to this unique subset of
individuals who have CKD (17,20). This article reviews the
contemporary aspects of care for these patients.
Kidney Allograft Function
Reduced kidney allograft function is associated directly with
poor graft and patient outcomes (9,10,15,16,21). A close fol-
low-up of kidney allograft function (monthly to every 2 mo in
stable cases) therefore is recommended in all of these patients.
Transplant recipients whose graft is stable after 2 yr may de-
crease the frequency of their actual visits to every 3 to 4 mo but
are encouraged to continue more frequent laboratory testing.
Traditionally, allograft function has been assessed by mea-
surement of serum creatinine values and urinalyses. The
Modification of Diet in Renal Disease (MDRD) estimation
formula for GFR in patients with CKD (22) provides a low
bias and high precision (2.7 and 10.4 ml/min per 1.73 m
2
,
respectively) in predicting GFR in kidney transplant recipi-
ents (23). The MDRD GFR calculator is available online at
http://www.kidney.org/professionals/kdoqi/index.cfm. The ad-
vantage of using GFR or creatinine clearance estimation formu-
las to determine the stage of CKD allows one to implement a
clinical action plan that is consistent with CKD guidelines (17).
Cystatin C is a novel and more sensitive marker of kidney
function in kidney transplant recipients that also may predict
successfully cardiovascular outcomes (2426). Whether one
uses the serum creatinine, cystatin C, or eGFR formula, it still is
crucial to screen for graft dysfunction at regular intervals. At no
time after transplantation can one assume that the allograft is
entirely stable (27). As with native kidney diseases, late allo-
graft dysfunction may be secondary to acute or chronic pro-
cesses.
Acute Kidney Injury or Failure
Acute kidney injury or kidney failure in the late posttrans-
plantation period may be divided into basic categories (Table
Figure 1. Impact of kidney transplant loss on patient survival.
Reprinted with permission from reference (10).
Table 1. Causes of late allograft dysfunction
a
Acute Renal Failure in the Late
Posttransplantation Period
Late Allograft Loss
Prerenal Chronic allograft dysfunction
decreased effective
circulating volume
CAN
CNI nephrotoxicity
volume contraction polyoma (BK) virus
congestive heart failure nephropathy
liver failure recurrent/de novo
renal transplant artery
stenosis
glomerular disease
chronic rejection
drugs (immunologic)
CNI
ACE-I, ARB
acute rejection
Patient death with
NSAID functioning graft
Renal CVD
urinary tract infection
and/or pyelonephritis
infectious complications
malignancies
acute rejection other
acute interstitial nephritis
acute tubular necrosis
recurrent/de novo
glomerular disease
Postrenal
hydronephrosis
a
ACE-I, angiotensin-converting enzyme inhibitor; ARB,
angiotensin receptor blocker; CAN, chronic allograft
nephropathy; CNI, calcineurin inhibitor; CVD, cardiovascular
disease; NSAID, nonsteroidal anti-inflammatory drugs.
624 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 623640, 2006
1). The principal difference is the possibility of acute rejection.
Therefore, the assessment and management of acute injury or
failure in these patients should include the history (including
adherence with immunosuppressive drugs) and physical exam-
ination, serum creatinine, blood urea nitrogen and electrolytes,
blood levels of calcineurin inhibitors (CNI) and/or sirolimus,
urinalysis, urine electrolytes, urine indices, and ultrasono-
graphic assessment of the allograft. Volume depletion, drugs,
and urinary tract infections constitute the largest proportion of
causes of acute injury or failure late after transplantation, and
management is similar to that undertaken in native kidney
disease. It is important to move readily toward treatment,
especially for urinary tract infections, because late infections
(after at least 12 mo of allograft function) are associated with a
reduction in graft survival (28). Acute rejection is a relatively
rare entity in this period. Its diagnosis requires a biopsy, pref-
erably at the transplant center, and should be considered when
the above diagnostics are negative or the creatinine fails to
return to the previous baseline. In general, any change in cre-
atinine 0.3 mg/dl (25 mol/L) needs to be evaluated
promptly.
Acute Rejection
Organ Procurement and Transplantation Network data from
April 2005 demonstrated that acute rejection rates within 1 yr
averaged 14.7% for the 2001 to 2003 period. After the first
posttransplant year, acute rejection rates are less prevalent. Not
all rejection episodes carry the same negative prognostic im-
pact. The available literature shows that recurrent rejection
(5,29), rejection episodes that occur in the setting of potent
immunosuppression (30), episodes that occur beyond month 6
(2932), or episodes with a lack of functional response to ther-
apy (5) have the worst impact on graft survival.
Chronic Allograft Dysfunction
Chronic allograft dysfunction after the first year posttrans-
plantation is secondary to chronic allograft nephropathy
(CAN), drug toxicity, recurrent and/or de novo glomerular
diseases, polyoma (BK) infections, and late acute rejection (Ta-
ble 1) (13,14).
CAN. CAN is a condition of altered kidney allograft func-
tion at least 3 mo posttransplantation, independent from acute
rejection, CNI toxicity, recurrent or de novo disease, or immu-
nologic features that are suggestive of chronic rejection (C4d

staining in the pericapillary tubules, intimal fibrosis with in-


flammatory cells, and duplication of glomerular basement
membrane) (3339). The term CAN is preferred to chronic
rejection because the latter implies an immunologic mechanism
of injury (33,37). However, immune and nonimmune features
(including chronic CNI toxicity) often coexist. Therefore, the
term CAN practically defines a histologic diagnosis that is
characterized by tubular atrophy, interstitial fibrosis, and fi-
brous intimal thickening with variable glomerular lesions (37
39). Accompanying clinical features include a gradual elevation
in serum creatinine, hypertension, and mild to moderate pro-
teinuria (1 to 3 g/24 h) (13,18). These nonspecific pathologic
findings result from a proinflammatory milieu in which the
level of injury from both immune and nonimmune insults
exceeds the capacity of tissue repair (13,38). CAN lesions are
present in up to 94% of grafts at 12 mo posttransplantation
(13,38,40) along with the noted clinical features.
CAN is associated with significant morbidity and mortality
and is the main reason for returning to dialysis after transplan-
tation (38). It is important to appreciate that early and late
posttransplantation events play a role in the progression of
CAN (Table 2).
CNI Nephrotoxicity. Cyclosporine A (CsA) and tacroli-
mus play a significant role in progressive kidney dysfunction
after transplantation (4043). Nearly all kidney allografts dis-
play histopathologic signs of chronic CNI toxicity by 10 yr
posttransplantation (40). CNI manifest nephrotoxicity in sev-
eral ways (41,44). They can mediate acute nephrotoxicity via
reversible afferent arteriolar vasoconstriction, often associated
with subtle volume depletion. This is a common cause for
transient increases in serum creatinine and needs to be fol-
lowed up in a timely manner after improved volume repletion.
CNI are causative agents of thrombotic microangiopathy and
can result independently in progressive and irreversible arte-
riolar hyaline changes and tubulointerstitial fibrosis (38,41,44).
A number of approaches have attempted to stave off nephro-
toxicity through CNI avoidance (4547), withdrawal (4854), or
minimization (51,5557). The rejection risk with any of these
strategies is relatively small (10%), and kidney allograft func-
tion may improve by up to 7 ml/min (5254). This should not
be attempted without previous consultation with the transplant
center.
Antibody-Mediated Rejection. Both cellular and humoral
processes contribute to the pathogenesis of chronic allograft
rejection (39). Antibody-mediated rejection (AMR) includes all
forms of rejection that involve donor-specific antibodies (DSA)
against HLA antigens, ABO isoagglutinins, or non-HLA anti-
gens (i.e., anti-endothelial antibodies) and is defined by the
presence of DSA in the recipients serum and C4d deposition in
the peritubular capillaries as a marker of antibody-mediated
Table 2. Factors involved in the development/
progression of CAN
a
Modifiable Risk
Factors
Nonmodifiable Risk
Factors
Nonimmune Nonimmune
CNI toxicity early delayed graft function
hypertension donor age or poor graft quality
dyslipidemia donor genetic factors
diabetes donor/recipient size mismatch
proteinuria Immune
oxidative stress HLA mismatch
chronic hypoxia previous sensitization (high PRA)
CMV infection previous rejection
Immune
chronic rejection
subclinical rejection
patient noncompliance
suboptimal
immunosuppression
a
CMV, cytomegalovirus; PRA, panel of reactive antigens.
Clin J Am Soc Nephrol 1: 623640, 2006 Medical Care of Kidney Transplant Recipients 625
activation of the complement cascade (58). The chronic form of
AMR occurs after the first year posttransplantation and is more
indolent than its early counterpart (58,59). It has been shown
that one third of patients with CAN develop de novo anti-HLA
antibodies compared with only 4% in kidney transplant recip-
ients with normal allograft function (60). Likewise, the presence
of C4d deposition in the peritubular capillaries has been iden-
tified in one third of patients with chronic allograft dysfunction
(61). Injury to peritubular capillaries can result progressively in
vascular dropout, interstitial fibrosis, and significant scarring of
the kidney allograft (62). Clinically, many of these patients
present with significant proteinuria that results from transplant
glomerulopathy, a specific form of glomerular lesion that is
considered to be the result of immune injury.
A large, multicenter, collaborative study of 2231 kidney
transplant recipients that was undertaken to determine the
frequency of anti-HLA antibodies in renal transplant recipients
with functional transplants (63) showed the presence of anti-
HLA antibodies in 20% of patients (63). In this study, 478
patients who developed anti-HLA antibodies had a signifi-
cantly higher rate of allograft loss at 2 yr than 1753 control
patients without detectable antibodies (15.1 versus 6.8%; P
0.0005) (64). Although these findings suggested a role for anti-
HLA antibodies as predictors of allograft loss, this predictive
value was NS for recipients with serum creatinine levels 2
mg/dl (177 mol/L) (64).
In summary, the current literature supports AMR as a con-
tributor to chronic allograft dysfunction through an indolent
and progressive process. Notwithstanding this evidence,
United Network for Organ Sharing data show that 23% of
HLA-identical kidneys are lost after 10 yr of transplantation,
demonstrating that other factorsincluding non-HLA antibod-
ies and nonimmunologic processesalso play an important
part in the pathogenesis of chronic allograft injury (65). Al-
though there is a role for the monitoring of HLA antibodies in
kidney transplant recipients, routine screening of DSA in kid-
ney transplant recipients is not currently recommended. High-
risk patients (patients with AMR, sensitized patients with PRA
80%, or patients who undergo desensitization protocols) with
increased serum creatinine levels (2 mg/dl) may benefit from
DSA screening and subsequent adjustment of immunosuppres-
sion. Many issues remain unsolved concerning the monitoring
of anti-HLA antibodies, including the choice of diagnostic tech-
niques (i.e., complement-based versus solid-phase assays), the
frequency of testing, and the interpretation of antibody data in
the absence of allograft dysfunction.
Recurrent and/or De Novo Disease. Glomerulonephritis
(GN) is responsible for kidney failure as a primary or contrib-
uting cause in 20 to 40% of patients who receive a transplant
(66). The incidence rate of GN recurrence posttransplantation
ranges between 6 and 19.4% and may result in graft loss in
approximately 8.4% of cases (6668). Because recurrence im-
plies that the patient has biopsy-proven GN in both native and
transplanted kidneys (a condition not always fulfilled), many
studies group de novo and recurrent GN (66). Considering these
limitations, any form of GN may recur at any time posttrans-
plantation (6668). However, focal and segmental glomerulo-
sclerosis, membranoproliferative GN (MPGN), hemolytic ure-
mic syndrome, and membranous glomerulonephritis (MGN)
are more likely to recur within the first posttransplant year,
whereas pauci-immune GN, IgA nephropathy, fibrillary GN,
lupus nephritis, and diabetes recur typically after the first year
(6668). Principal causes of de novo GN in renal allografts
include transplant glomerulopathy, hemolytic uremic syn-
drome, membranous glomerulonephritis, and MPGN (69). The
last two have been associated with hepatitis C virus (HCV)
infection, although MPGN (with or without cryoglobulinemia)
is more commonly described (70,71). HCV infection may be
associated more frequently with glomerular disease in the renal
allograft compared with native kidneys possibly because of
increased HCV RNA titers that result from immunosuppres-
sion (72). Recurrent/de novo GN is associated overall with a
two-fold increased risk for graft loss (6668). It therefore is
important to suspect this condition in individuals with protein-
uria, hematuria, and a slow decline in allograft function after
the first year. The diagnostic approach should include a biopsy
with electron microscopy and immunohistology studies in ad-
dition to appropriate serologic evaluation (6668).
Treatment options for recurrent/de novo GN remain limited
and tend to be supportive, especially after the first posttrans-
plant year. Because these patients are already on immunosup-
pressive agents, management focuses on the removal of the
underlying cause if identified (infection, drugs, or malignancy)
and improved control of BP, proteinuria, and dyslipidemia. The
benefits of angiotensin-converting enzyme inhibitors (ACE-I)
and angiotensin receptor blockers (ARB) in native kidney dis-
easemediated proteinuria have not been demonstrated as ex-
tensively in the transplant population, yet these agents have
important antiproteinuric effects that can improve allograft
survival in individuals with significant proteinuria (73). Cyto-
toxic medications such as cyclophosphamide may have limited
benefits for specific posttransplantation entities, e.g., MPGN
(74), recurrent antiglomerular basement membrane glomeru-
lonephritis (75), and focal and segmental glomerulosclerosis
(76), but their use has to be gauged carefully and other anti-
proliferative immunosuppressive drug dosages may have to be
altered to decrease untoward effects. When graft loss ensues,
retransplantation remains an option. However, the risk for
recurrence typically increases with the number of transplants
(67,77).
Polyoma Virus Infection. Polyoma viruses are ubiquitous
DNA viruses that infect a variety of animals, including hu-
mans. BK virus, a human polyoma virus, was described ini-
tially in a transplant recipient (11,78). It has a remarkable
tropism for the genitourinary tract. BK virusinduced nephrop-
athy is recognized as an important cause of late allograft fail-
ure. The reported incidence of allograft failure has ranged from
15 to 50% in affected individuals (11,7981). Unfortunately,
there are no clinical features that are unique to BK virus infec-
tion. There often is an unexplained rise in serum creatinine,
asymptomatic hematuria, ureteral stenosis, and/or subacute
kidney failure as a result of interstitial nephritis (11,7981).
Diagnosis can be made by biopsy and urine cytology with
identification of characteristic cytopathologic changes. Addi-
626 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 623640, 2006
tional data suggest that assessment and monitoring of serum
PCR for viral DNA and viral load may be helpful as an adjunct
diagnostic and monitoring test (11,7982). Unfortunately, spe-
cific antiviral therapy for BK nephritis does not currently exist.
Clinical trials have suggested efficacy with low-dose cidofovir
(0.25 to 0.33 mg/kg every 2 to 3 wk intravenously) (81,83) as
well as leflunomide (83,84). Because BK virus infection reflects
the overall state of immunosuppression regardless of the choice
of CNI or adjuvant therapy (82), the basis of treatment is to
decrease immunosuppressive medications (11,7984). This is
another excellent example of the importance of transplant bi-
opsy to investigate an otherwise unexplained, persistent in-
crease in the serum creatinine.
Proteinuria
The evaluation of kidney function in kidney transplant re-
cipients should include routine measurement of urinary protein
excretion. Urine protein to creatinine ratios are a reliable sub-
stitute for 24-h urine collection (85). Proteinuria may be defined
as 200 mg/g creatinine and microalbuminuria 30 mg/g
creatinine on spot urine samples (86). The incidence of protein-
uria in kidney transplant patients ranges between 10 and 25%
(27). Proteinuria from the native kidneys normally resolves
within the first 1 to 10 wk posttransplantation (85). The three
most common causes of persistent proteinuria after kidney
transplantation are CAN, recurrent glomerulonephritis, and
drug-related nephrotoxicity (27,68,8789). Renal vein thrombo-
sis and reflux nephropathy are less common causes of protein-
uria after transplantation (Table 3).
Proteinuria reflects the severity of the underlying glomerular
and tubulointerstitial injury. Presumably, proteinuria also con-
tributes to ongoing transplant dysfunction and fibrosis through
putative mechanisms that involve aberrant proximal tubule
protein uptake and tubular cell toxicity (90,91). It is striking that
despite the evidence that demonstrates the significance of pro-
teinuria in native kidneymediated disease, only recently has
proteinuria been examined in depth in kidney transplantation.
In general, it is an independent risk factor for disease progres-
sion, graft failure, and patient death after kidney transplanta-
tion (27,8690,92). It is interesting that proteinuria may be a
clinical surrogate for predicting outcomes in conjunction with
medication changes. Urinary protein excretion 800 mg/d is
the only independent predictor of negative outcomes in pa-
tients with chronic allograft dysfunction, converted from CNI
to sirolimus (50). Transplant patients therefore should be eval-
uated regularly for proteinuria every 6 to 12 mo after the first
year posttransplantation (27). Because of the cost-effectiveness
of these laboratory tests, we suggest that a spot urine protein/
creatinine be obtained along with a urinalysis at each actual
clinic visit.
Several studies suggest that ACE-I and ARB can have a
beneficial effect in prolonging allograft survival, especially in
transplant recipients with CAN and proteinuria (73,9395). It is
important to strive for BP control (130/80 mmHg and poten-
tially lower if proteinuria is 1 g/24 h) (96) and to look for
possible adverse effects when using these agents. Although up
to a 30% increase in serum creatinine levels after the initiation
of antiproteinuric therapy may be acceptable (97), careful mon-
itoring of kidney function, hematocrit (Hct), and serum potas-
sium levels is warranted as these drugs may result in acute
kidney failure, anemia, and hyperkalemia in kidney transplant
recipients (14).
Cardiovascular and Peripheral Vascular
Disease
It is important to recognize that these entities are intertwined
in terms of pathogenesis and outcomes. CVD is the principal
cause of death in kidney transplant recipients (Figure 2)
(9,10,12,96,98,99). Mortality related to cardiovascular causes ac-
counts for nearly one quarter (23%) of all-cause mortality by 15
yr posttransplantation (100). Of note, even a nonfatal posttrans-
plantation myocardial infarction may predict future graft fail-
ure and death (101). Peripheral vascular disease (cerebral vas-
cular disease and lower extremity vascular disease) afflicts at
least 15% of all kidney transplant recipients in a 10- to 15-yr
period posttransplantation (100).
Both traditional and nontraditional cardiovascular risk fac-
tors, including the concurrent presence of peripheral vascular
disease, contribute to CVD morbidity and mortality in kidney
transplant recipients (Table 4) (96,102). The NKF K/DOQI work
group on CVD concluded that modifiable risk factors should be
targeted for intervention in these patients (Table 4) (96). Tradi-
tional CVD risk factors are shared risk factors for both CKD and
CVD. Therefore, addressing each should have a dual impact.
Table 3. Proteinuria after kidney transplantation
Common causes
CAN
recurrent/de novo disease
native disease (usually resolved by third month
posttransplantation)
urinary tract infections
Less common causes
renal transplant vein thrombosis
renal transplant artery stenosis
drugs
reflux
Figure 2. Cardiovascular mortality in kidney transplant recipi-
ents. Reprinted with permission from reference (98).
Clin J Am Soc Nephrol 1: 623640, 2006 Medical Care of Kidney Transplant Recipients 627
Symptomatic patients should undergo prompt functional
and/or invasive testing even if angiography is required. Intra-
venous hydration with bicarbonate (103) or saline, oral N-ace-
tylcysteine (104), and/or ascorbic acid (105) and withholding
CNI, ACE-I, and ARB for 12 h before angiography likely will
reduce the risk for contrast-induced nephropathy in the allo-
graft. Currently, there is insufficient evidence to recommend
for or against routine periodic screening for asymptomatic
CVD in kidney transplant recipients (27). However, we believe
that individuals who are at high-risk, e.g., two or more tradi-
tional cardiac risk factors or a history of a pretransplantation
cardiovascular event or a positive pretransplantation screening
test may benefit from noninvasive stress imaging every 1 to 2 yr
after transplantation.
Hypertension
Both the seventh report of the Joint National Committee on
Prevention, Detection, Evaluation, and Treatment of high BP
and the K/DOQI clinical practice guidelines on hypertension
and antihypertensive agents in CKD defined normal BP as
120/80 mmHg (97,106). With this definition, the incidence of
posttransplantation hypertension ranges up to 80%. The causes
of posttransplantation hypertension are varied and include
CNI use, prednisone, preexisting hypertension, primary kidney
disease, kidney transplant artery stenosis, and graft dysfunc-
tion (100,107). Systolic and diastolic hypertension both are in-
dependent risk factors for graft and patient loss in kidney
transplant recipients (108110). Surprisingly, no large, random-
ized, controlled studies have demonstrated improved long-
term outcomes by decreasing BP in kidney transplantation, yet
a recent analysis of the Collaborative Transplant Study showed
that lower systolic BP (SBP), even after the first posttransplant
year, was associated with improved graft and patient survival
(111). Patients whose SBP was 140 mmHg at 1 yr posttrans-
plantation but controlled to 140 mmHg by 3 yr had signifi-
cantly improved long-term graft outcome compared with pa-
tients with sustained high SBP (relative risk 0.79; 95%
confidence interval 0.73 to 0.86; P 0.001) (111).
As part of best medical practice, it therefore is appropriate to
treat posttransplantation hypertension to target BP values
whenever possible (27,97,106,112). Both the seventh report of
the Joint National Committee and K/DOQI guidelines recom-
mended a target BP of 130/80 mmHg in patients with CKD,
although only K/DOQI specifically addresses BP targets in
renal transplant recipients (97,106). The American Society of
Transplantation (an older guideline) recommends target BP
levels of 140/90 mmHg (27), and the EBPG recommend man-
datory BP control to levels of 130/85 mmHg in kidney trans-
plant patients without proteinuria and 125/75 mmHg in pro-
teinuric patients (112).
Lifestyle modifications are necessary and should include
weight reduction, a DASH (Dietary Approaches to Stop Hyper-
tension) eating plan, dietary sodium reduction, and physical
activity (97). All classes of antihypertensive drugs have been
used in transplant patients, yet no preferred agents are offered
by any of the guidelines. Initial efficacy studies suggested that
calcium channel blockers might have greater benefit in achiev-
ing BP control and limiting graft loss (107,113), but it has not
been shown that calcium channel blockers have a clear benefit
over ACE-I on long-term kidney allograft function and survival
(112). Furthermore, these drugs may be involved in the devel-
opment of edema and CsA-induced gingival hyperplasia (114).
On the basis of the effect of ACE-I and ARB in slowing the rate
of progression in patients with CKD, several groups have used
these agents in CAN, proteinuria, and underlying heart disease
posttransplantation (73,9395,115117). These studies have dis-
sipated initial concerns regarding the safety of ACE-I and ARB
because no significant hyperkalemia or acute changes in kidney
function were observed. However, caution is advised in indi-
viduals with preexisting hyperkalemia, anemia, and suspected
transplant renal artery stenosis (118). The use of diuretics in
hypertensive renal transplant patients may be effective and
often is necessary in combination therapies. These drugs may
result in hypercalcemia, hyponatremia, hyperuricemia, and im-
paired glucose/lipid metabolism in a high-risk group of pa-
tients.
In summary, a target BP of 130/80 mmHg with lower BP
levels (125/75 mmHg if proteinuria 1 g/d) is recommended
in renal transplant recipients (Figure 3). ACE-I and ARB may be
used safely as preferred agents in patients with proteinuria
(urine protein:creatinine ratio 200 mg/g), tolerating up to a
35% rise in baseline serum creatinine levels. Combination ther-
apy frequently is required to control posttransplantation hyper-
tension (96,97,106,110); regardless of the agents selected, a close
follow-up (weekly for 2 wk, then monthly) of BP, serum creat-
inine, electrolytes, and hemoglobin (Hb) levels (in case of
ACE-I and ARB use) is recommended. It is advised to avoid
making changes in BP medications concomitant (within 1 wk)
with changes in immunosuppression. This may complicate the
differential diagnosis in case of a rapid rise in serum creatinine
levels.
Dyslipidemia
Dyslipidemia, alone or as part of the metabolic syndrome, is an
established risk factor for CVD mortality in kidney transplant
recipients (27,119121), as it is in the general population. Post-
Table 4. Traditional and nontraditional risk factors for
CVD in renal transplant recipients
Traditional Risk
Factors
Nontraditional Risk
Factors
Older age Decreased kidney function
Male gender CNI
Family history of CVD Proteinuria
Diabetes Anemia
Hypertension C-reactive protein
Dyslipidemia Oxidative stress
low HDL Advanced glycation end products
high LDL Inflammation
Physical inactivity Homocysteine
Left ventricular
hypertrophy
Uric acid
Hyperparathyroidism
Menopause Obesity
Tobacco use Thrombogenic factors
628 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 623640, 2006
transplantation hyperlipidemia affects 60 to 80% of patients (122)
and is associated with immunosuppressive treatment, including
sirolimus, corticosteroids, and CsA (121123). Other factors, such
as preexisting familial dyslipidemia, obesity, kidney dysfunction,
proteinuria, and diabetes, also contribute to posttransplantation
hyperlipidemia (121,124). Retrospective studies have reported
posttransplantation dyslipidemia as a risk factor for long-term
graft loss (125). However, the Assessment of Lescol in Renal
Transplant (ALERT) trial, the only randomized, double-blind, pla-
cebo-controlled study to examine the role of a statin on renal end
points, found no significant effect of fluvastatin on kidney trans-
plant outcomes (126). In a post hoc analysis of this trial, fluvastatin
significantly decreased the incidence of myocardial infarction and
cardiac death (127). Other studies have shown similar findings in
that statins can improve patient survival by an average of 24%
among kidney transplant recipients (128).
The evaluation and treatment of posttransplantation dyslip-
idemia should be based on the National Cholesterol Education
Program III and the NKF K/DOQI clinical practice guidelines
(119,121). Dyslipidemia in kidney transplant recipients should
be evaluated at baseline, 2 to 3 mo after a change in treatment,
and at least annually thereafter (121). Recommended goals are
target LDL, non-HDL cholesterol, and triglyceride levels of
100, 130, and 150 mg/dl, respectively (121). Treatment op-
tions include modification of the immunosuppressive regimen
when possible, therapeutic lifestyle changes, statins, fibrates,
nicotinic acid, and bile acid sequestrants (121).
All statins can provide a 30 to 40% reduction in LDL choles-
terol levels. Patients who require maximal LDL cholesterol
lowering may be treated with atorvastatin or simvastatin,
whereas patients with low HDL cholesterol may have a greater
advantage from simvastatin use and those with elevated trig-
lycerides may benefit from high-dose atorvastatin. The final
choice of statins should be left to the judgment of the treating
physician. For the treatment of hypertriglyceridemia, particu-
larly in patients who are treated with sirolimus, gemfibrozil
may be the fibric derivative of choice (121). Although nicotinic
acid derivatives also could be used, fibric acid derivatives are
better tolerated (121). Bile acid sequestrants such as cholestyra-
mine, colestipol, and colesevelam hydrochloride alter the bio-
availability of immunosuppressants and also may increase tri-
glyceride levels (121). Ezetimibe was used recently alone and in
combination with statins to reduce LDL cholesterol in a small-
size study with renal transplant patients (129). However, larger
clinical trials are required to establish the safety and the efficacy
of this drug in kidney transplantation. Antilipemic drugs may
have significant interactions between the classes and with im-
munosuppressant agents. Statins and fibrates interact with CNI
and may result in hepatitis, myositis, and rhabdomyolysis
(121,124). The prevalence of these untoward effects is minimal
in our contemporary era of immunosuppression in the absence
of high-dose fibrate or statin therapy and with close monitoring.
In summary, adult renal transplant recipients with hypertri-
glyceridemia (triglycerides 300 mg/dl, or 5.65 mmol/L) may
be treated with fibrates, and statins can be used for LDL cho-
lesterol levels of 100 mg/dl (2.59 mmol/L; Figure 4). Treat-
ment of proteinuria and other causes of secondary dyslipide-
mia, along with therapeutic lifestyle changes including diet and
physical activity, always should be combined. It monitoring of
treatment efficacy and observing for clinical and biologic signs
of myositis and hepatitis with most current antilipemic agents
are recommended.
Diabetes
New-onset diabetes after transplantation (NODAT) is de-
fined using the definition of diabetes and impaired glucose
tolerance by the World Health Organization and the American
Diabetes Association (www.diabetes.org) (130,131). Fasting
plasma glucose levels 126 mg/dl (7 mmol/L) or 2-h plasma
glucose levels 200 mg/dl (11.1 mmol/L) during an oral glu-
cose tolerance test defines diabetes (130,131). NODAT has a
high incidence in kidney transplant recipients (up to 25%) and
significantly decreases patient and graft survival (132135), and
its prevalence increases continuously with time. Immunosup-
pressive therapy with tacrolimus, older recipient age, deceased-
donor status, hepatitis C, rejection episodes, black race, and
higher body weight all are independent risk factors for NODAT
(Table 5) (132,135,136).
Figure 3. BP management after the first posttransplant year.
Figure 4. Dyslipidemia management after the first posttrans-
plant year.
Clin J Am Soc Nephrol 1: 623640, 2006 Medical Care of Kidney Transplant Recipients 629
The International Consensus Guidelines on New-Onset Dia-
betes after Transplantation recommended management strate-
gies to reduce onset and complications are available on the
International Diabetes Federation web site (http://www.
idf.org) (131). Regular evaluation for diabetic retinopathy, ne-
phropathy, and neuropathy should be performed, and appro-
priate measures should be taken. Therapy for all kidney trans-
plant recipients with diabetes should be directed at achievable
targets, including BP 130/80 mmHg, fasting plasma glucose
126 mg/dl, glycosylated Hb 6.5g/dl, urine protein:creati-
nine ratio 200 mg/g, and LDL 100 mg/dl (131,137).
In summary, NODAT has been identified as one of the most
important factors associated with reduced graft function and
patient and graft survival. International Consensus Guidelines
on New-onset Diabetes after Transplantation recommend a
multifaceted approach of weight control, diet and exercise, oral
agent mono- or combination therapy and insulin use for tight
glycemic control (Figure 5) (131). Sulfonylureas, meglitinides,
biguanides, thiazolidinediones, and -glucosidase inhibitors all
may be used depending on the individual characteristics of the
patient and risk for possible side effects (Figure 5) (131,137).
Proteinuria, dyslipidemia, hypertension, and anemia should be
treated accordingly.
Anemia
The American Society of Transplantation defines anemia as
Hb levels of 13 g/dl (Hct of 42%) for male patients and 12
g/dl (Hct of 37%) for female patients (27). The NKF K/DOQI
clinical practice guidelines define anemia as Hb levels 12 g/dl
(Hct of 37%) for male patients and 11 g/dl (Hct of 33%) for
premenopausal women and prepubertal patients (138). Anemia
is a common complication after kidney transplantation, with an
incidence approximating 30 to 40% (139141). It may result
from inadequate erythropoietin production; iron deficiency;
malignancies; oxidative stress/inflammation; hemolytic and
uremic syndrome; and viral infections, including CMV and
Parvovirus B19 infections (Figure 6) (139141). Drugs such as
sirolimus, mycophenolate mofetil, azathioprine, ACE-I, and
ARB are the principal agents that are responsible for late post-
transplantation anemia (118,139142). Although late (1 yr
posttransplantation) anemia has been reported as an indepen-
dent risk factor for cardiovascular morbidity and mortality in
this patient population (143145), there is insufficient evidence
at present as to whether aggressive anemia management can
improve outcomes. Pending the results of prospective, random-
ized trials to determine optimal Hb/Hct levels in kidney trans-
plant recipients, the K/DOQI recommendations for the man-
agement of anemia in patients with CKD may be the best
guideline (138).
In the late posttransplantation period, Hb/Hct levels should
be checked monthly. Anemia workup should be started for Hb
levels 12 g/dl, and treatment with erythropoietic-stimulating
agents (ESA) should be started for Hb 11 g/dl. Target recom-
mended Hb (Hct) should be the same as in patients with CKD
(138), i.e., between 11 (33%) and 12 g/dl (36%), pending the
results of prospective, randomized trials. The current recom-
mended subcutaneous dose of epoetin- in dialysis patients is
80 to 120 IU/kg per wk in two to three divided doses (138). In
renal transplant recipients with anemia, ESA may be dosed
Table 5. Risk factors for NODAT
a
Recipient characteristics
older age (45 yr)
higher body mass index (30)
black race
family history of diabetes
Hispanic ethnicity
education (no college degree)
Donor
deceased donor
male gender
Transplant era (after 1995)
Tacrolimus use
HLA mismatch
Acute rejection
HCV infection
a
HCV, hepatitis C virus; NODAT, new-onset diabetes after
transplantation.
Figure 5. Diabetes management after the first posttransplant
year.
Figure 6. Common causes of posttransplantation anemia. MMF,
mycophenolate mofetil; AZA, azathioprine; CMV, cytomegalo-
virus; TMA, thrombotic microangiopathy.
630 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 623640, 2006
weekly and their dose changed monthly if needed. If the rise in
Hct after ESA therapy has been 2% during a 2- to 4-wk
period, then the ESA dose should be increased by 50% (138).
Conversely, if Hct levels increase 8% or if the Hb/Hct exceeds
the target Hb/Hct, then the weekly dose should be decreased
by 25%. Abrupt discontinuation of ESA therapy may result in a
collapse of erythropoiesis. Iron supplementation (oral/intrave-
nous) is required for transferrin saturation 20% and/or fer-
ritin levels 100 ng/ml (138).
In summary, posttransplantation anemia is a prevalent com-
plication and an independent risk factor for cardiovascular
morbidity and mortality in kidney transplant recipients. Ane-
mia workup should be initiated for Hb levels 12 g/dl, and
treatment should be targeted to a goal Hb between 11 and 12
g/dl. This goal may be achieved by the removal of causal
agents (Figure 6), oral/intravenous iron supplementation, and
ESA (138,146149).
Bone Metabolism and Disease
Almost all kidney transplant recipients have some degree of
osteodystrophy (disorders of bone remodeling and modeling)
that persists after kidney transplantation (27,150153). The in-
cidence of osteoporosis (bone mineral density 2.5 SD below
the young adult mean value or T score) approximates 60%, and
it results in significant morbidity and mortality, including avas-
cular necrosis and fractures (27,150,151). Maximum bone loss
occurs within the first 3 to 6 mo posttransplantation and con-
tinues during the later stages at a slower rate. Etiologic features
include treatment with corticosteroids and CNI, previous os-
teodystrophy, persistent hyperparathyroidism, hypogonadism,
metabolic acidosis, and smoking (150,151).
Current management recommendations are based on the
available clinical trials and extrapolation from findings in pa-
tients with CKD (151,152,154,155). The cornerstone of these
recommendations is regular follow-up of serum calcium, phos-
phorus, and intact parathyroid hormone levels and annual (at
least during the first 3 yr posttransplantation) assessment of
bone mineral density with dual energy x-ray absorptiometry
scan of the lumbar spine and the dominant hip. Treatment
strategies to reduce bone loss include testosterone/estrogen
replacement when appropriate, elemental calcium and vitamin
D, phosphate binders, correction of metabolic acidosis, weight-
bearing exercise, and oral or intravenous bisphosphonates
(151,152,154,155). Although these strategies result in the pre-
vention of bone loss, a recent meta-analysis of clinical trials that
used bisphosphonates, vitamin D analogues, calcitonin, and
hormone replacement therapy showed no benefit from any
intervention to reduce the incidence of fractures in kidney
transplant recipients (156). Despite recommendations for the
early use of bisphosphonates (152), these antiresorptive agents
present a clear risk for adynamic bone disease (157) and should
be used cautiously in patients with impaired kidney function
after the first year.
The management of tertiary hyperparathyroidism (HPTH)
after transplantation also may be challenging. The EBPG for
renal transplantation recommend that individuals with tertiary
HPTH be observed for 1 year after transplantation whenever
possible to allow for a spontaneous involution (158). After the
first year, persistent HPTH may be treated by surgical parathy-
roidectomy (159), although this procedure may result in de-
creased kidney allograft function through mechanisms that
remain to be defined (160,161). Recently, calcimimetic cinacal-
cet was used to normalize posttransplantation hypercalcemia in
patients with persistent HPTH (162,163). This drug may present
a reasonable alternative to surgical parathyroidectomy. How-
ever, its safety and effectiveness need to be examined in larger
clinical trials.
Infections
Infections remain the second most common cause of death in
kidney transplant recipients (9,10,12,13,27,164). The risk for
infection in these patients is determined primarily by the in-
tensity of exposure to potential pathogens and the net state of
immunosuppression (165). Overall, the most prevalent oppor-
tunistic infections are viral, and CMV is the primary virus
involved. However, a host of community-acquired and oppor-
tunistic bacterial, viral, and fungal infections may occur at
different rates depending on the period after transplantation.
After the first posttransplant year, the majority of patients
benefit from good allograft function and are maintained on
minimal immunosuppression. Community-acquired respira-
tory infections are the principal cause of infectious complica-
tions, as in the general population. Opportunistic infections are
uncommon in this setting except when there is intense expo-
sure to environmental pathogens (165). In a group of patients
with chronic, progressive viral infections (CMV, Epstein-Barr
virus, papillomavirus, HBV, or HCV), signs of progressive
organ damage may be observed (liver or skin cancer or retinitis
with CMV). Finally, a small proportion of individuals may
receive increased immunosuppression for acute rejection epi-
sodes. These patients are at high risk for severe opportunistic
infections, including Pneumocystis carinii, Listeria monocytogenes,
Nocardia asteroides, Cryptococcus neoformans, and aspergillus
(165).
To reduce the burden of infection-related morbidity and
mortality, the American Society of Transplantation has devel-
oped infectious disease guidelines for the prevention and man-
agement of infectious complications of solid-organ transplan-
tation (166). In addition, guidelines on vaccinations, drug
interactions with anti-infective agents, and strategies for safe
living after organ transplantation are available on the American
Society of Transplantation web site (http://www.a-s-t.org/
mobile/index.htm). Briefly, patient vaccination status should
be reviewed at the first clinic visit, and a vaccination strategy
should be developed, keeping in mind that live vaccines are not
given after transplantation and that patients, close contacts, and
family members should receive injectable influenza vaccine
yearly (inhaled influenza vaccine should not be given to trans-
plant recipients or family members). Pneumococcal polysaccha-
ride vaccine should be administered before transplantation and
repeated every 3 to 5 yr after initial vaccination. Vaccination
series should be restarted approximately 6 mo after transplan-
tation, and efficacy should be documented by serologic assays
when available. Finally, adult travelers after kidney transplan-
Clin J Am Soc Nephrol 1: 623640, 2006 Medical Care of Kidney Transplant Recipients 631
tation need appropriate counseling and vaccinations before
their trip (167).
In brief, infection and rejection are the two ends of the net
state of immunosuppression in kidney transplant recipients. A
primary goal is the prevention and/or effective treatment of
infection. After the first year posttransplantation, most com-
mon infections are community-acquired respiratory infections.
In subgroups of patients who have chronic viral infections or
are receiving increased immunosuppression for late rejection
episodes, there is greater risk for malignancies and opportunis-
tic infections. Close monitoring of these patients is required for
early diagnosis of complications. Preventive strategies are nec-
essary and encompass counseling and vaccinations.
Malignancies
Posttransplantation malignancies are a major cause of mor-
tality in kidney transplant recipients (10,14,99). These individ-
uals have a significantly increased risk for malignancies com-
pared with age-matched healthy control subjects and even
patients who have ESRD and are on the waiting list for a
transplant (27,168170) (Figure 7). General and more trans-
plant-specific factors contribute to the increased incidence of
malignancies in this patient population. These include age,
smoking, immunosuppression, and chronic viral infections
(169171).
The relative risk for cancer depends on the type of malig-
nancy. It ranges from a two- to three-fold increase for common
malignancies such as lung, prostate, breast, and colon to up to
a 100-fold increase for entities such as Kaposis sarcomas, post-
transplantation lymphoproliferative disease and nonmelano-
matous skin cancers (168170) (Figure 7).
After the first posttransplant year, kidney transplant recipi-
ents should undergo annual or biannual skin examination.
Age-appropriate annual prostate-specific antigen measure-
ments, fecal occult blood testing, digital and rectal examina-
tions, breast examination and mammography, and colonoscopy
are indicated as in the nontransplant patient. Unique screening
may include hepatobiliary ultrasound and serum -fetoprotein
measurements (history of HBV or HCV) as well as cystoscopy
(history of cyclophosphamide use; Table 6) (27,169). In addition
to specific antitumor therapy, treatment strategies for post-
transplantation malignancies include decreasing or modifying
the immunosuppressive regimen (169,172,173). The antiprolif-
erative characteristics of sirolimus may have a salutary impact
on the treatment of some of these malignancies. In a recent
report, regression of cutaneous Kaposis sarcoma was observed
after a switch from a CsA mycophenolate mofetil regimen to
a regimen based on sirolimus in 15 kidney transplant recipients
(173). Sirolimus use has also been associated with decreased
incidence of cancer (including skin cancers) in the first 2 yr
posttransplantation (174).
In summary, with the growing number of kidney transplant
recipients, the prevention, early diagnosis, and treatment of
posttransplantation malignancies remain an important chal-
lenge to both the transplant and the primary care communities
(Figure 7, Table 6). The development of any malignancy should
mandate contact with the transplant center to coordinate the
overall approach to the patient, including tapering of immuno-
suppression.
Obesity and Metabolic Syndrome
Overweight and obesity, defined by a body mass index 25
and 30 kg/m
2
, respectively, present a major quandary after
kidney transplantation (159). Similar to the trend in the general
population, 60% of current kidney transplant recipients are
overweight or obese at the time of transplantation, representing
a 116% increase from 1987 (175). A significant number of pa-
tients gain additional weight after transplantation: The average
weight gain in the first posttransplant year was reported re-
cently to be approximately 3 kg (176). Both posttransplantation
weight gain and obesity are associated with decreased long-
term allograft and patient outcomes (176180). Although it
Figure 7. Posttransplantation malignancies. EBV, Epstein-Barr virus; HPV, human papillomavirus; HBV, hepatitis B virus; HBC,
hepatitis C virus; NHL, non-Hodgkins lymphoma; Skin (NM), skin cancer (nonmelanomas).
632 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 623640, 2006
remains unknown whether it is the higher incidence of cardio-
vascular comorbid conditions that affects graft outcomes or
obesity-specific issues (hyperfiltration and glomerulopathy),
most studies report a higher likelihood of premature death and
graft loss in obese kidney transplant recipients (176181). Fe-
male, young, black, low-income patients with type 2 diabetes
are at the highest risk for posttransplantation obesity (175,182).
The EBPG considers obesity as a risk factor for increased CVD
after transplantation and recommends lifestyle measures to
address the epidemic (179). However, this complex problem
requires a multifaceted approach and a combination of diet,
exercise, minimization of steroid therapy, and psychologic sup-
port often is needed. Gastric bypass surgery may be a safe and
effective means for achieving significant long-term weight loss
and relief of comorbid conditions after transplantation
(183,184). This approach should be reserved for resistant cases
and requires close collaboration with the transplant center.
The metabolic syndrome (MS) is a condition of impaired
glucose and insulin metabolism, overweight and abdominal fat
distribution, mild dyslipidemia, and hypertension. It is preva-
lent in the US population (age-adjusted prevalence 25%) and is
associated with subsequent development of type 2 diabetes and
CVD (185,186). The working definition of the National Choles-
terol Education Program Expert Panel Adult Treatment Panel
III defines MS as a condition with three or more of the follow-
ing: (1) Waist circumference 102 cm in men and 88 cm in
women, (2) serum triglyceride level 150 mg/dl (1.69 mmol/L),
(3) HDL cholesterol level 40 mg/dl (1.04 mmol/L) in men
and 50 mg/dl (1.29 mmol/L) in women, (4) BP 130/85
mmHg, and (5) serum glucose level 110 mg/dl (6.1 mmol/L)
(187). Data to address the role of MS in kidney transplantation
are scarce (120,188,189). It seems that up to 63% of renal trans-
plant recipients experience MS at a median time of 6 yr post-
transplantation (120). Obesity is a risk factor for developing MS
in kidney transplant patients (188), and waist circumference has
been found to be an independent determinant of inflammation
measured by C-reactive protein levels in these patients (189). A
recent study showed that MS is associated with impaired kid-
ney allograft function after the first posttransplant year, but
only SBP and hypertriglyceridemia seemed to be associated
independently with the decline in kidney function (120).
In summary, weight gain, obesity, and MS are prevalent after
the first posttransplant year. Although there is an association
between these conditions and decreased long-term patient and
allograft outcomes, it remains unknown whether there is un-
derlying causality. A multifaceted approach that includes a
combination of diet, exercise, minimization of steroid therapy,
psychologic support, and gastric bypass surgery may be re-
quired to address this growing epidemic in renal transplant
patients.
Quality of Life and Nonadherence
Kidney transplantation produces greater improvements in
quality of life (QOL) than does dialysis (190,191). A meta-
analysis of prospective studies that evaluated QOL before and
after kidney transplantation found consistent improvement
among the posttransplantation population (192). When specific
domains of QOL were examined, 78% of the studies found
improvement in physical functioning, 85% in mental health,
and 63% in social functioning (192). However, despite such
evidence, many kidney transplant recipients experience ad-
verse effects that affect life satisfaction and QOL (193195).
Sixty-six percent of kidney transplant recipients reported easy
bruising/slow wound healing, 61% noted adverse effects re-
lated to sexuality, and 50% related problems secondary to
changes in physical appearance (193,194).
Transplant recipients are required to take immunosup-
pressive treatment for the rest of their lives, and, unfortu-
nately, adherence to this regimen often is suboptimal. This
may have dramatic effects on graft and patient outcomes
because up to 91% of nonadherent patients may experience
graft loss or death (196). A recent meta-analysis of 325 stud-
ies that were published from 1980 to 2001 and reported the
frequency and impact of nonadherence in adult renal trans-
plant recipients showed that a median (interquartile range)
of 22% (18 to 26%) of recipients were nonadherent and that
the odds for graft failure increased seven-fold (95% confi-
dence interval 4 to 12%) in nonadherent patients compared
with adherent patients (197). Psychosocial and financial bar-
riers often contribute to poor adherence with medications;
however, these are not the only factors: A 1-yr posttrans-
Table 6. Screening for malignancies after the first year posttransplantation
a
Screening Frequency 30 Days 6 Months 12 Months 5 Years
Skin and lip Self-exam Physical
Anogenital Physical, pelvic
PAP smear biopsy
Uterine cervix Pelvic, PAP smear
Sarcomas (including Kaposis) High-risk ethnic
groups
Skin, conjunctiva, oropharyngeal
mucosa biopsy
PTLD Self-exam Physical
Hepatic (HCV and HBV patients) -fetoprotein
Breast Self-exam Physical, mammogram
Colorectal Colonoscopy
Prostate Rectal, PSA
a
HBV, hepatitis B virus; PAP, Papanicolaou; PSA, prostate-specific antigen; PTLD, posttransplantation lymphoproliferative
disease.
Clin J Am Soc Nephrol 1: 623640, 2006 Medical Care of Kidney Transplant Recipients 633
plantation compliance rate of only 48% was reported in a free
multidrug immunosuppressive clinic (198).
Conclusions
Kidney transplantation is the optimal treatment for patients
with ESRD; however, despite improvements in short-term pa-
tient and graft outcomes, there has been no major improvement
in long-term outcomes. There is a need for collaboration among
the transplant center, community nephrologists, and primary
care physicians who are involved in the long-term care of these
patients to enhance these outcomes. A number of public re-
sources that provide clinical practice guidelines for the treat-
ment of these patients are available on the Internet (Table 7).
Prevention and early management of disease progression, car-
diovascular complications, infections, and malignancies consti-
tute the cornerstone of this collaborative effort to extend life
span and allograft function.
Acknowledgments
Parts of this work were supported by an American Society of Trans-
plantation/Fujisawa Fellowship Award (A.D.); American Heart Asso-
ciation grant 0235290N (M.S.); and National Institutes of Health grants
KO8-DK067981 (A.D.), RO1-AI49285-05 (B.N.B.), and K24-DK616962-04
(B.N.B.).
References
1. Wolfe RA, Ashby VB, Milford EL, Ojo AO, Ettenger RE,
Agodoa LY, Held PJ, Port FK: Comparison of mortality in
all patients on dialysis, patients on dialysis awaiting trans-
plantation, and recipients of a first cadaveric transplant.
N Engl J Med 341: 17251730, 1999
2. Sayegh MH, Carpenter CB: Transplantation 50 years later:
Progress, challenges, and promises. N Engl J Med 351:
27612766, 2004
3. Kaplan B, Meier-Kriesche HU: Renal transplantation: A
half century of success and the long road ahead. J Am Soc
Nephrol 15: 32703271, 2004
4. USRDS 2004 annual data report. Am J Kidney Dis 45: 8280,
2005
5. Meier-Kriesche HU, Schold JD, Srinivas TR, Kaplan B: Lack
of improvement in renal allograft survival despite a
marked decrease in acute rejection rates over the most
recent era. Am J Transplant 4: 378383, 2004
6. Meier-Kriesche HU, Schold JD, Kaplan B: Long-term renal
allograft survival: Have we made significant progress or is
it time to rethink our analytic and therapeutic strategies?
Am J Transplant 4: 12891295, 2004
7. Keith DS, DeMattos A, Golconda M, Prather J, Cantarovich
M, Paraskevas S, Tchervenkov J, Norman DJ: Factors asso-
ciated with improvement in deceased donor renal allograft
function in the 1990s. J Am Soc Nephrol 16: 15121521, 2005
8. Hariharan S, Johnson CP, Bresnahan BA, Taranto SE,
McIntosh MJ, Stablein D: Improved graft survival after
renal transplantation in the United States, 1988 to 1996.
N Engl J Med 342: 605612, 2000
9. Meier-Kriesche HU, Baliga R, Kaplan B: Decreased renal
function is a strong risk factor for cardiovascular death
after renal transplantation. Transplantation 75: 12911295,
2003
10. Kaplan B, Meier-Kriesche HU: Death after graft loss: An
important late study endpoint in kidney transplantation.
Am J Transplant 2: 970974, 2002
11. Mannon RB: Polyomavirus nephropathy: What have we
learned? Transplantation 77: 13131318, 2004
12. Ojo AO, Hanson JA, Wolfe RA, Leichtman AB, Agodoa LY,
Port FK: Long-term survival in renal transplant recipients
with graft function. Kidney Int 57: 307313, 2000
13. Pascual M, Theruvath T, Kawai T, Tolkoff-Rubin N, Co-
simi AB: Strategies to improve long-term outcomes after
renal transplantation. N Engl J Med 346: 580590, 2002
14. Djamali A, Premasathian N, Pirsch JD: Outcomes in kidney
transplantation. Semin Nephrol 23: 306316, 2003
15. Karthikeyan V, Karpinski J, Nair RC, Knoll G: The burden
of chronic kidney disease in renal transplant recipients.
Am J Transplant 4: 262269, 2004
16. Djamali A, Kendziorski C, Brazy PC, Becker BN: Disease
progression and outcomes in chronic kidney disease and
renal transplantation. Kidney Int 64: 18001807, 2003
17. Levey AS, Coresh J, Balk E, Kausz AT, Levin A, Steffes
MW, Hogg RJ, Perrone RD, Lau J, Eknoyan G: National
Kidney Foundation practice guidelines for chronic kidney
disease: Evaluation, classification, and stratification. Ann
Intern Med 139: 137147, 2003
18. Gourishankar S, Hunsicker LG, Jhangri GS, Cockfield SM,
Table 7. Useful Internet resources
American Diabetes Association (ADA) http://www.diabetes.org/homepage.jsp
American Society of Nephrology (ASN) http://ww.asn-online.org
American Society of Transplantation (AST) http://www.a-s-t.org
European Best Practice Guidelines (EBPG) http://www.ndt-educational.org/guidelines.asp
International Diabetes Federation (IDF) http://www.idf.org
Kidney Disease: Improving Global Outcomes (KDIGO) http://kdigo.org/welcome.htm
National Kidney Foundation (NKF) http://www.kidney.org
NKF Clinical Practice Guidelines http://www.kidney.org/professionals/kdoqi/
guidelines/cfm
Scientific Registry of Transplant Recipients (SRTR) http://www.ustransplant.org
United Network for Organ Sharing (UNOS) http://www.unos.org
United States Renal Data System (USRDS) http://www.usrds.org
634 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 623640, 2006
Halloran PF: The stability of the glomerular filtration rate
after renal transplantation is improving. J Am Soc Nephrol
14: 23872394, 2003
19. Kasiske BL, Gaston RS, Gourishankar S, Halloran PF, Ma-
tas AJ, Jeffery J, Rush D: Long-term deterioration of kidney
allograft function. Am J Transplant 5: 14051414, 2005
20. Levey AS, Eckardt KU, Tsukamoto Y, Levin A, Coresh J,
Rossert J, Zeeuw D, Hostetter TH, Lameire N, Eknoyan G:
Definition and classification of chronic kidney disease: A
position statement from Kidney Disease: Improving Global
Outcomes (KDIGO). Kidney Int 67: 20892100, 2005
21. Hariharan S, McBride MA, Cherikh WS, Tolleris CB,
Bresnahan BA, Johnson CP: Post-transplant renal function
in the first year predicts long-term kidney transplant sur-
vival. Kidney Int 62: 311318, 2002
22. Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D:
A more accurate method to estimate glomerular filtration
rate from serum creatinine: A new prediction equation.
Modification of Diet in Renal Disease Study Group. Ann
Intern Med 130: 461470, 1999
23. Gaspari F, Ferrari S, Stucchi N, Centemeri E, Carrara F,
Pellegrino M, Gherardi G, Gotti E, Segoloni G, Salvadori
M, Rigotti P, Valente U, Donati D, Sandrini S, Sparacino V,
Remuzzi G, Perico N: Performance of different prediction
equations for estimating renal function in kidney trans-
plantation. Am J Transplant 4: 18261835, 2004
24. Hariharan S, Kasiske B, Matas A, Cohen A, Harmon W,
Rabb H: Surrogate markers for long-term renal allograft
survival. Am J Transplant 4: 11791183, 2004
25. Levin A: Cystatin C, serum creatinine, and estimates of
kidney function: Searching for better measures of kidney
function and cardiovascular risk. Ann Intern Med 142: 586
588, 2005
26. Gokkusu CA, Ozden TA, Gul H, Yildiz A: Relationship
between plasma cystatin C and creatinine in chronic renal
diseases and Tx-transplant patients. Clin Biochem 37: 9497,
2004
27. Kasiske BL, Vazquez MA, Harmon WE, Brown RS, Dano-
vitch GM, Gaston RS, Roth D, Scandling JD, Singer GG:
Recommendations for the outpatient surveillance of renal
transplant recipients. American Society of Transplantation.
J Am Soc Nephrol 11[Suppl 15]: S1S86, 2000
28. Abbott KC, Swanson SJ, Richter ER, Bohen EM, Agodoa
LY, Peters TG, Barbour G, Lipnick R, Cruess DF: Late
urinary tract infection after renal transplantation in the
United States. Am J Kidney Dis 44: 353362, 2004
29. Leggat JE Jr, Ojo AO, Leichtman AB, Port FK, Wolfe RA,
Turenne MN, Held PJ: Long-term renal allograft survival:
Prognostic implication of the timing of acute rejection ep-
isodes. Transplantation 63: 12681272, 1997
30. Meier-Kriesche HU, Ojo AO, Hanson JA, Cibrik DM,
Punch JD, Leichtman AB, Kaplan B: Increased impact of
acute rejection on chronic allograft failure in recent era.
Transplantation 70: 10981100, 2000
31. Basadonna GP, Matas AJ, Gillingham KJ, Payne WD, Dunn
DL, Sutherland DE, Gores PF, Gruessner RW, Najarian JS:
Early versus late acute renal allograft rejection: Impact on
chronic rejection. Transplantation 55: 993995, 1993
32. Nett PC, Heisey DM, Shames BD, Fernandez LA, Pirsch JD,
Sollinger HW: Influence of kidney function to the impact of
acute rejection on long-term kidney transplant survival.
Transpl Int 18: 385389, 2005
33. Solez K, Axelsen RA, Benediktsson H, Burdick JF, Cohen
AH, Colvin RB, Croker BP, Droz D, Dunnill MS, Halloran
PF: International standardization of criteria for the histo-
logic diagnosis of renal allograft rejection: The Banff work-
ing classification of kidney transplant pathology. Kidney Int
44: 411422, 1993
34. Racusen LC, Solez K, Colvin RB, Bonsib SM, Castro MC,
Cavallo T, Croker BP, Demetris AJ, Drachenberg CB, Fogo
AB, Furness P, Gaber LW, Gibson IW, Glotz D, Goldberg
JC, Grande J, Halloran PF, Hansen HE, Hartley B, Hayry
PJ, Hill CM, Hoffman EO, Hunsicker LG, Lindblad AS,
Yamaguchi Y: The Banff 97 working classification of renal
allograft pathology. Kidney Int 55: 713723, 1999
35. Racusen LC, Halloran PF, Solez K: Banff 2003 meeting
report: New diagnostic insights and standards. Am J Trans-
plant 4: 15621566, 2004
36. Racusen LC, Colvin RB, Solez K, Mihatsch MJ, Halloran
PF, Campbell PM, Cecka MJ, Cosyns JP, Demetris AJ,
Fishbein MC, Fogo A, Furness P, Gibson IW, Glotz D,
Hayry P, Hunsickern L, Kashgarian M, Kerman R, Magil
AJ, Montgomery R, Morozumi K, Nickeleit V, Randhawa
P, Regele H, Seron D, Seshan S, Sund S, Trpkov K: Anti-
body-mediated rejection criteria: An addition to the Banff
97 classification of renal allograft rejection. Am J Transplant
3: 708714, 2003
37. Colvin RB: Chronic allograft nephropathy. N Engl J Med
349: 22882290, 2003
38. Halloran PF, Melk A, Barth C: Rethinking chronic allograft
nephropathy: The concept of accelerated senescence. J Am
Soc Nephrol 10: 167181, 1999
39. Joosten SA, Sijpkens YW, van Kooten C, Paul LC: Chronic
renal allograft rejection: Pathophysiologic considerations.
Kidney Int 68: 113, 2005
40. Nankivell BJ, Borrows RJ, Fung CL, OConnell PJ, Allen
RD, Chapman JR: The natural history of chronic allograft
nephropathy. N Engl J Med 349: 23262333, 2003
41. Bennett WM, DeMattos A, Meyer MM, Andoh T, Barry JM:
Chronic cyclosporine nephropathy: The Achilles heel of
immunosuppressive therapy. Kidney Int 50: 10891100,
1996
42. Halloran PF: Immunosuppressive drugs for kidney trans-
plantation. N Engl J Med 351: 27152729, 2004
43. Ojo AO, Held PJ, Port FK, Wolfe RA, Leichtman AB,
Young EW, Arndorfer J, Christensen L, Merion RM:
Chronic renal failure after transplantation of a nonrenal
organ. N Engl J Med 349: 931940, 2003
44. Burdmann EA, Andoh TF, Yu L, Bennett WM: Cyclospor-
ine nephrotoxicity. Semin Nephrol 23: 465476, 2003
45. Tran HT, Acharya MK, McKay DB, Sayegh MH, Carpenter
CB, Auchincloss H Jr, Kirkman RL, Milford EL: Avoidance
of cyclosporine in renal transplantation: Effects of dacli-
zumab, mycophenolate mofetil, and steroids. J Am Soc
Nephrol 11: 19031909, 2000
46. Flechner SM, Kurian SM, Solez K, Cook DJ, Burke JT,
Rollin H, Hammond JA, Whisenant T, Lanigan CM, Head
SR, Salomon DR: De novo kidney transplantation without
use of calcineurin inhibitors preserves renal structure and
function at two years. Am J Transplant 4: 17761785, 2004
47. Knechtle SJ, Pirsch JD, Fechner H, Becker BN, Friedl A,
Colvin RB, Lebeck LK, Chin LT, Becker YT, Odorico JS,
DAlessandro AM, Kalayoglu M, Hamawy MM, Hu H,
Bloom DD, Sollinger HW: Campath-1H induction plus
Clin J Am Soc Nephrol 1: 623640, 2006 Medical Care of Kidney Transplant Recipients 635
rapamycin monotherapy for renal transplantation: Results
of a pilot study. Am J Transplant 3: 722730, 2003
48. Kreis H, Oberbauer R, Campistol JM, Mathew T, Daloze P,
Schena FP, Burke JT, Brault Y, Gioud-Paquet M, Scarola JA,
Neylan JF: Long-term benefits with sirolimus-based ther-
apy after early cyclosporine withdrawal. J Am Soc Nephrol
15: 809817, 2004
49. Mota A, Arias M, Taskinen EI, Paavonen T, Brault Y,
Legendre C, Claesson K, Castagneto M, Campistol JM,
Hutchison B, Burke JT, Yilmaz S, Hayry P, Neylan JF:
Sirolimus-based therapy following early cyclosporine
withdrawal provides significantly improved renal histol-
ogy and function at 3 years. Am J Transplant 4: 953961,
2004
50. Diekmann F, Budde K, Oppenheimer F, Fritsche L, Neu-
mayer HH, Campistol JM: Predictors of success in conver-
sion from calcineurin inhibitor to sirolimus in chronic al-
lograft dysfunction. Am J Transplant 4: 18691875, 2004
51. Weir MR, Ward MT, Blahut SA, Klassen DK, Cangro CB,
Bartlett ST, Fink JC: Long-term impact of discontinued or
reduced calcineurin inhibitor in patients with chronic allo-
graft nephropathy. Kidney Int 59: 15671573, 2001
52. Kasiske BL, Chakkera HA, Louis TA, Ma JZ: A meta-
analysis of immunosuppression withdrawal trials in renal
transplantation. J Am Soc Nephrol 11: 19101917, 2000
53. Mulay AV, Hussain N, Fergusson D, Knoll GA: Cal-
cineurin inhibitor withdrawal from sirolimus-based ther-
apy in kidney transplantation: A systematic review of ran-
domized trials. Am J Transplant 5: 17481756, 2005
54. Johnson RW, Kreis H, Oberbauer R, Brattstrom C, Claesson
K, Eris J: Sirolimus allows early cyclosporine withdrawal
in renal transplantation resulting in improved renal func-
tion and lower blood pressure. Transplantation 72: 777786,
2001
55. Weir MR, Anderson L, Fink JC, Gabregiorgish K,
Schweitzer EJ, Hoehn-Saric E, Klassen DK, Cangro CB,
Johnson LB, Kuo PC, Lim JY, Bartlett ST: A novel approach
to the treatment of chronic allograft nephropathy. Trans-
plantation 64: 17061710, 1997
56. Pilmore HL, Dittmer ID: Calcineurin inhibitor nephrotox-
icity: Reduction in dose results in marked improvement in
renal function in patients with coexisting chronic allograft
nephropathy. Clin Transplant 16: 191195, 2002
57. Weir MR, Blahut S, Drachenburg C, Young C, Papademi-
triou J, Klassen DK, Cangro CB, Bartlett ST, Fink JC: Late
calcineurin inhibitor withdrawal as a strategy to prevent
graft loss in patients with suboptimal kidney transplant
function. Am J Nephrol 24: 379386, 2004
58. Montgomery RA, Hardy MA, Jordan SC, Racusen LC,
Ratner LE, Tyan DB, Zachary AA: Consensus opinion from
the antibody working group on the diagnosis, reporting,
and risk assessment for antibody-mediated rejection and
desensitization protocols. Transplantation 78: 181185, 2004
59. Terasaki PI, Cai J: Humoral theory of transplantation: Fur-
ther evidence. Curr Opin Immunol 17: 541545, 2005
60. Poggio ED, Clemente M, Riley J, Roddy M, Greenspan NS,
Dejelo C, Najafian N, Sayegh MH, Hricik DE, Heeger PS:
Alloreactivity in renal transplant recipients with and with-
out chronic allograft nephropathy. J Am Soc Nephrol 15:
19521960, 2004
61. Regele H, Bohmig GA, Habicht A, Gollowitzer D, Schill-
inger M, Rockenschaub S, Watschinger B, Kerjaschki D,
Exner M: Capillary deposition of complement split product
C4d in renal allografts is associated with basement mem-
brane injury in peritubular and glomerular capillaries: A
contribution of humoral immunity to chronic allograft re-
jection. J Am Soc Nephrol 13: 23712380, 2002
62. Ishii Y, Sawada T, Kubota K, Fuchinoue S, Teraoka S,
Shimizu A: Injury and progressive loss of peritubular cap-
illaries in the development of chronic allograft nephropa-
thy. Kidney Int 67: 321332, 2005
63. Terasaki PI, Ozawa M: Predicting kidney graft failure by
HLA antibodies: A prospective trial. Am J Transplant 4:
438443, 2004
64. Terasaki PI, Ozawa M: Predictive value of HLA antibodies
and serum creatinine in chronic rejection: Results of a
2-year prospective trial. Transplantation 80: 11941197, 2005
65. Terasaki PI: Deduction of the fraction of immunologic and
non-immunologic failure in cadaver donor transplants.
Clin Transpl 449452, 2003
66. Chadban S: Glomerulonephritis recurrence in the renal
graft. J Am Soc Nephrol 12: 394402, 2001
67. Briganti EM, Russ GR, McNeil JJ, Atkins RC, Chadban SJ:
Risk of renal allograft loss from recurrent glomerulone-
phritis. N Engl J Med 347: 103109, 2002
68. Hariharan S, Adams MB, Brennan DC, Davis CL, First MR,
Johnson CP, Ouseph R, Peddi VR, Pelz CJ, Roza AM,
Vincenti F, George V: Recurrent and de novo glomerular
disease after renal transplantation: A report from Renal
Allograft Disease Registry (RADR). Transplantation 68: 635
641, 1999
69. Denton MD, Singh AK: Recurrent and de novo glomeru-
lonephritis in the renal allograft. Semin Nephrol 20: 164
175, 2000
70. Cruzado JM, Gil-Vernet S, Ercilla G, Seron D, Carrera M,
Bas J, Torras J, Alsina J, Grinyo JM: Hepatitis C virus-
associated membranoproliferative glomerulonephritis in
renal allografts. J Am Soc Nephrol 7: 24692475, 1996
71. Morales JM, Campistol JM: Transplantation in the patient
with hepatitis C. J Am Soc Nephrol 11: 13431353, 2000
72. Periera BJ, Wright TL, Schmid CH, Levey AS: The impact
of pretransplantation hepatitis C infection on the outcome
of renal transplantation. Transplantation 60: 799805, 1995
73. Premasathian NC, Muehrer R, Brazy PC, Pirsch JD, Becker
BN: Blood pressure control in kidney transplantation:
Therapeutic implications. J Hum Hypertens 18: 871877,
2004
74. Lien YH, Scott K: Long-term cyclophosphamide treatment
for recurrent type I membranoproliferative glomerulone-
phritis after transplantation. Am J Kidney Dis 35: 539543,
2000
75. European best practice guidelines for renal transplanta-
tion. Section IV: Long-term management of the transplant
recipient. IV. 2.5. Chronic graft dysfunction. Late recur-
rence of primary glomerulonephritides. Nephrol Dial Trans-
plant 17[Suppl 4]: 1618, 2002
76. Newstead CG: Recurrent disease in renal transplants.
Nephrol Dial Transplant 18[Suppl 6]: vi68vi74, 2003
77. Briggs JD, Jones E: Recurrence of glomerulonephritis fol-
lowing renal transplantation. Scientific Advisory Board of
the ERA-EDTA Registry. European Renal Association-Eu-
ropean Dialysis and Transplant Association. Nephrol Dial
Transplant 14: 564565, 1999
78. Gardner SD, Field AM, Coleman DV, Hulme B: New hu-
636 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 623640, 2006
man papovavirus (B.K.) isolated from urine after renal
transplantation. Lancet 1: 12531257, 1971
79. Fishman JA: BK virus nephropathy: Polyomavirus adding
insult to injury. N Engl J Med 347: 527530, 2002
80. Hirsch HH, Knowles W, Dickenmann M, Passweg J, Klim-
kait T, Mihatsch MJ, Steiger J: Prospective study of poly-
omavirus type BK replication and nephropathy in renal-
transplant recipients. N Engl J Med 347: 488496, 2002
81. Hirsch HH, Brennan DC, Drachenberg CB, Ginevri F, Gor-
don J, Limaye AP, Mihatsch MJ, Nickeleit V, Ramos E,
Randhawa P, Shapiro R, Steiger J, Suthanthiran M, Trofe J:
Polyomavirus-associated nephropathy in renal transplan-
tation: Interdisciplinary analyses and recommendations.
Transplantation 79: 12771286, 2005
82. Brennan DC, Agha I, Bohl DL, Schnitzler MA, Hardinger
KL, Lockwood M, Torrence S, Schuessler R, Roby T, Gau-
dreault-Keener M, Storch GA: Incidence of BK with tacroli-
mus versus cyclosporine and impact of preemptive immu-
nosuppression reduction. Am J Transplant 5: 582594, 2005
83. Farasati NA, Shapiro R, Vats A, Randhawa P: Effect of
leflunomide and cidofovir on replication of BK virus in an
in vitro culture system. Transplantation 79: 116118, 2005
84. Williams JW, Javaid B, Kadambi PV, Gillen D, Harland R,
Thistlewaite JR, Garfinkel M, Foster P, Atwood W, Millis
JM, Meehan SM, Josephson MA: Leflunomide for poly-
omavirus type BK nephropathy. N Engl J Med 352: 1157
1158, 2005
85. DCunha PT, Parasuraman R, Venkat KK: Rapid resolution
of proteinuria of native kidney origin following live donor
renal transplantation. Am J Transplant 5: 351355, 2005
86. Eknoyan G, Hostetter T, Bakris GL, Hebert L, Levey AS,
Parving HH, Steffes MW, Toto R: Proteinuria and other
markers of chronic kidney disease: A position statement of
the national kidney foundation (NKF) and the national
institute of diabetes and digestive and kidney diseases
(NIDDK). Am J Kidney Dis 42: 617622, 2003
87. Barnas U, Schmidt A, Haas M, Kaider A, Tillawi S, Wamser
P, Mayer G: Parameters associated with chronic renal
transplant failure. Nephrol Dial Transplant 12[Suppl 2]: 82
85, 1997
88. Roodnat JI, Mulder PG, Rischen-Vos J, van Riemsdijk IC,
van Gelder T, Zietse R, IJzermans JN, Weimar W: Protein-
uria after renal transplantation affects not only graft sur-
vival but also patient survival. Transplantation 72: 438444,
2001
89. Fernandez-Fresnedo G, Plaza JJ, Sanchez-Plumed J, Sanz-
Guajardo A, Palomar-Fontanet R, Arias M: Proteinuria: A
new marker of long-term graft and patient survival in
kidney transplantation. Nephrol Dial Transplant 19[Suppl 3]:
iii47iii51, 2004
90. Zandi-Nejad K, Eddy AA, Glassock RJ, Brenner BM: Why
is proteinuria an ominous biomarker of progressive kidney
disease? Kidney Int Suppl S76S89, 2004
91. Zoja C, Morigi M, Remuzzi G: Proteinuria and phenotypic
change of proximal tubular cells. J Am Soc Nephrol 14[Suppl
1]: S36S41, 2003
92. Rosenkranz AR, Mayer G: Proteinuria in the transplanted
patient. Nephrol Dial Transplant 15: 12901292, 2000
93. Artz MA, Hilbrands LB, Borm G, Assmann KJ, Wetzels JF:
Blockade of the renin-angiotensin system increases graft
survival in patients with chronic allograft nephropathy.
Nephrol Dial Transplant 19: 28522857, 2004
94. Lin J, Valeri AM, Markowitz GS, DAgati VD, Cohen DJ,
Radhakrishnan J: Angiotensin converting enzyme inhibi-
tion in chronic allograft nephropathy. Transplantation 73:
783788, 2002
95. Remuzzi G, Perico N: Routine renin-angiotensin system
blockade in renal transplantation? Curr Opin Nephrol Hy-
pertens 11: 110, 2002
96. Levey AS: Controlling the epidemic of cardiovascular dis-
ease in chronic renal disease: Where do we start? Am J
Kidney Dis 32[Suppl 3]: S5S13, 1998
97. Chobanian AV, Bakris GL, Black HR, Cushman WC, Green
LA, Izzo JL Jr, Jones DW, Materson BJ, Oparil S, Wright JT
Jr, Roccella EJ: The Seventh Report of the Joint National
Committee on Prevention, Detection, Evaluation, and
Treatment of High Blood Pressure: The JNC 7 report.
JAMA 289: 25602572, 2003
98. Foley RN, Parfrey PS, Sarnak MJ: Clinical epidemiology of
cardiovascular disease in chronic renal disease. Am J Kid-
ney Dis 32[Suppl 3]: S112S119, 1998
99. Briggs JD: Causes of death after renal transplantation.
Nephrol Dial Transplant 16: 15451549, 2001
100. Kasiske BL, Guijarro C, Massy ZA, Wiederkehr MR, Ma JZ:
Cardiovascular disease after renal transplantation. J Am
Soc Nephrol 7: 158165, 1996
101. Lentine KL, Brennan DC, Schnitzler MA: Incidence and
predictors of myocardial infarction after kidney transplan-
tation. J Am Soc Nephrol 16: 496506, 2005
102. Ducloux D, Kazory A, Chalopin JM: Predicting coronary
heart disease in renal transplant recipients: A prospective
study. Kidney Int 66: 441447, 2004
103. Merten GJ, Burgess WP, Gray LV, Holleman JH, Roush TS,
Kowalchuk GJ, Bersin RM, Van Moore A, Simonton CA
3rd, Rittase RA, Norton HJ, Kennedy TP: Prevention of
contrast-induced nephropathy with sodium bicarbonate: A
randomized controlled trial. JAMA 291: 23282334, 2004
104. Tepel M, van der GM, Schwarzfeld C, Laufer U, Liermann
D, Zidek W: Prevention of radiographic-contrast-agent-
induced reductions in renal function by acetylcysteine.
N Engl J Med 343: 180184, 2000
105. Spargias K, Alexopoulos E, Kyrzopoulos S, Iacovis P,
Greenwood DC, Manginas A, Voudris V, Pavlides G,
Buller CE, Kremastinos D, Cokkinos DV: Ascorbic acid
prevents contrast-mediated nephropathy in patients with
renal dysfunction undergoing coronary angiography or
intervention. Circulation 110: 28372842, 2004
106. K/DOQI clinical practice guidelines on hypertension and
antihypertensive agents in chronic kidney disease. Am J
Kidney Dis 43: S1S230, 2004
107. Midtvedt K, Hartmann A: Hypertension after kidney
transplantation: Are treatment guidelines emerging? Neph-
rol Dial Transplant 17: 11661169, 2002
108. Opelz G, Wujciak T, Ritz E: Association of chronic kidney
graft failure with recipient blood pressure. Collaborative
Transplant Study. Kidney Int 53: 217222, 1998
109. Mange KC, Cizman B, Joffe M, Feldman HI: Arterial hy-
pertension and renal allograft survival. JAMA 283: 633
638, 2000
110. Kasiske BL, Anjum S, Shah R, Skogen J, Kandaswamy C,
Danielson B, OShaughnessy EA, Dahl DC, Silkensen JR,
Sahadevan M, Snyder JJ: Hypertension after kidney trans-
plantation. Am J Kidney Dis 43: 10711081, 2004
111. Opelz G, Dohler B: Improved long-term outcomes after
Clin J Am Soc Nephrol 1: 623640, 2006 Medical Care of Kidney Transplant Recipients 637
renal transplantation associated with blood pressure con-
trol. Am J Transplant 5: 27252731, 2005
112. European best practice guidelines for renal transplanta-
tion. Section IV: Long-term management of the transplant
recipient. IV. 5.2. Cardiovascular risks. Arterial hyperten-
sion. Nephrol Dial Transplant 17[Suppl 4]: 2526, 2002
113. Midtvedt K, Hartmann A, Foss A, Fauchald P, Nordal KP,
Rootwelt K, Holdaas H: Sustained improvement of renal
graft function for two years in hypertensive renal trans-
plant recipients treated with nifedipine as compared to
lisinopril. Transplantation 72: 17871792, 2001
114. Morales JM, Rodriguez-Paternina E, Araque A, Andres A,
Hernandez E, Ruilope LM, Rodicio JL: Long-term protec-
tive effect of a calcium antagonist on renal function in
hypertensive renal transplant patients on cyclosporine
therapy: A 5-year prospective randomized study. Trans-
plant Proc 26: 25982599, 1994
115. Maschio G, Alberti D, Janin G, Locatelli F, Mann JF, Mo-
tolese M, Ponticelli C, Ritz E, Zucchelli P: Effect of the
angiotensin-converting-enzyme inhibitor benazepril on the
progression of chronic renal insufficiency. The Angioten-
sin-Converting-Enzyme Inhibition in Progressive Renal In-
sufficiency Study Group. N Engl J Med 334: 939945, 1996
116. Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA,
Lewis JB, Ritz E, Atkins RC, Rohde R, Raz I: Renoprotec-
tive effect of the angiotensin-receptor antagonist irbesartan
in patients with nephropathy due to type 2 diabetes.
N Engl J Med 345: 851860, 2001
117. Tutone VK, Mark PB, Stewart GA, Tan CC, Rodger RS,
Geddes CC, Jardine AG: Hypertension, antihypertensive
agents and outcomes following renal transplantation. Clin
Transplant 19: 181192, 2005
118. Winkelmayer WC, Kewalramani R, Rutstein M, Gabardi S,
Vonvisger T, Chandraker A: Pharmacoepidemiology of
anemia in kidney transplant recipients. J Am Soc Nephrol
15: 13471352, 2004
119. Executive Summary of The Third Report of The National
Cholesterol Education Program (NCEP) Expert Panel on
Detection, Evaluation, and Treatment of High Blood Cho-
lesterol in Adults (Adult Treatment Panel III). JAMA 285:
24862497, 2001
120. de Vries AP, Bakker SJ, van Son WJ, van der Heide JJ, Ploeg
RJ, The HT, de Jong PE, Gans RO: Metabolic syndrome is
associated with impaired long-term renal allograft func-
tion; not all component criteria contribute equally. Am J
Transplant 4: 16751683, 2004
121. K/DOQI clinical practice guidelines for managing dyslipi-
demias in kidney transplant patients. Am J Transplant
4[Suppl 7]: S7S53, 2004
122. Kobashigawa JA, Kasiske BL: Hyperlipidemia in solid or-
gan transplantation. Transplantation 63: 331338, 1997
123. Brattstrom C, Wilczek H, Tyden G, Bottiger Y, Sawe J,
Groth CG: Hyperlipidemia in renal transplant recipients
treated with sirolimus (rapamycin). Transplantation 65:
12721274, 1998
124. Abtahi P, Zand MS: Management of hyperlipidemia in the
stable solid organ transplant recipient. Graft 4: 266274,
2001
125. Wissing KM, Abramowicz D, Broeders N, Vereerstraeten
P: Hypercholesterolemia is associated with increased kid-
ney graft loss caused by chronic rejection in male patients
with previous acute rejection. Transplantation 70: 464472,
2000
126. Fellstrom B, Holdaas H, Jardine AG, Holme I, Nyberg G,
Fauchald P, Gronhagen-Riska C, Madsen S, Neumayer
HH, Cole E, Maes B, Ambuhl P, Olsson AG, Hartmann A,
Logan JO, Pedersen TR: Effect of fluvastatin on renal end
points in the Assessment of Lescol in Renal Transplant
(ALERT) trial. Kidney Int 66: 15491555, 2004
127. Jardine AG, Holdaas H, Fellstrom B, Cole E, Nyberg G,
Gronhagen-Riska C, Madsen S, Neumayer HH, Maes B,
Ambuhl P, Olsson AG, Holme I, Fauchald P, Gimpelwicz
C, Pedersen TR: Fluvastatin prevents cardiac death and
myocardial infarction in renal transplant recipients: Post-
hoc subgroup analyses of the ALERT Study. Am J Trans-
plant 4: 988995, 2004
128. Cosio FG, Pesavento TE, Pelletier RP, Henry M, Ferguson
RM, Kim S, Lemeshow S: Patient survival after renal trans-
plantation. III: The effects of statins. Am J Kidney Dis 40:
638643, 2002
129. Puthenparumpil JJ, Keough-Ryan T, Kiberd M, Lawen J,
Kiberd BA: Treatment of hypercholesterolemia with
ezetimibe in the kidney transplant population. Transplant
Proc 37: 10331035, 2005
130. Report of the expert committee on the diagnosis and clas-
sification of diabetes mellitus. Diabetes Care 26[Suppl 1]:
S5S20, 2003
131. Davidson J, Wilkinson A, Dantal J, Dotta F, Haller H,
Hernandez D, Kasiske BL, Kiberd B, Krentz A, Legendre C,
Marchetti P, Markell M, van der Woude FJ, Wheeler DC:
New-onset diabetes after transplantation: 2003 Interna-
tional consensus guidelines. Proceedings of an interna-
tional expert panel meeting. Barcelona, Spain, 19 February
2003. Transplantation 75[Suppl]: SS3SS24, 2003
132. Cosio FG, Pesavento TE, Kim S, Osei K, Henry M, Fergu-
son RM: Patient survival after renal transplantation: IV.
Impact of post-transplant diabetes. Kidney Int 62: 1440
1446, 2002
133. Vesco L, Busson M, Bedrossian J, Bitker MO, Hiesse C,
Lang P: Diabetes mellitus after renal transplantation: Char-
acteristics, outcome, and risk factors. Transplantation 61:
14751478, 1996
134. Miles AM, Sumrani N, Horowitz R, Homel P, Maursky V,
Markell MS, Distant DA, Hong JH, Sommer BG, Friedman
EA: Diabetes mellitus after renal transplantation: As dele-
terious as non-transplant-associated diabetes? Transplanta-
tion 65: 380384, 1998
135. Kasiske BL, Snyder JJ, Gilbertson D, Matas AJ: Diabetes
mellitus after kidney transplantation in the United States.
Am J Transplant 3: 178185, 2003
136. Heisel O, Heisel R, Balshaw R, Keown P: New onset dia-
betes mellitus in patients receiving calcineurin inhibitors:
A systematic review and meta-analysis. Am J Transplant 4:
583595, 2004
137. Wilkinson A, Davidson J, Dotta F, Home PD, Keown P,
Kiberd B, Jardine A, Levitt N, Marchetti P, Markell M,
Naicker S, OConnell P, Schnitzler M, Standl E, Torregosa
JV, Uchida K, Valantine H, Villamil F, Vincenti F, Wissing
M: Guidelines for the treatment and management of new-
onset diabetes after transplantation. Clin Transplant 19:
291298, 2005
138. IV. NKF-K/DOQI clinical practice guidelines for anemia of
638 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 623640, 2006
chronic kidney disease: Update 2000. Am J Kidney Dis 37:
S182S238, 2001
139. Yorgin PD, Scandling JD, Belson A, Sanchez J, Alexander
SR, Andreoni KA: Late post-transplant anemia in adult
renal transplant recipients. An under-recognized problem?
Am J Transplant 2: 429435, 2002
140. Vanrenterghem Y, Ponticelli C, Morales JM, Abramowicz
D, Baboolal K, Eklund B, Kliem V, Legendre C, Morais
Sarmento AL, Vincenti F: Prevalence and management of
anemia in renal transplant recipients: A European survey.
Am J Transplant 3: 835845, 2003
141. Pirsch JD, Becker BN: Another cause for the same old
problem. Am J Transplant 4: 19311932, 2004
142. Augustine JJ, Knauss TC, Schulak JA, Bodziak KA, Siegel
C, Hricik DE: Comparative effects of sirolimus and myco-
phenolate mofetil on erythropoiesis in kidney transplant
patients. Am J Transplant 4: 20012006, 2004
143. Djamali A, Becker YT, Simmons WD, Johnson CA, Prema-
sathian N, Becker BN: Increasing hematocrit reduces early
posttransplant cardiovascular risk in diabetic transplant
recipients. Transplantation 76: 816820, 2003
144. Rigatto C, Foley R, Jeffery J, Negrijn C, Tribula C, Parfrey
P: Electrocardiographic left ventricular hypertrophy in re-
nal transplant recipients: Prognostic value and impact of
blood pressure and anemia. J Am Soc Nephrol 14: 462468,
2003
145. Rigatto C, Parfrey P, Foley R, Negrijn C, Tribula C, Jeffery
J: Congestive heart failure in renal transplant recipients:
Risk factors, outcomes, and relationship with ischemic
heart disease. J Am Soc Nephrol 13: 10841090, 2002
146. Chandler G, Harchowal J, Macdougall IC: Intravenous iron
sucrose: Establishing a safe dose. Am J Kidney Dis 38:
988991, 2001
147. Becker BN, Becker YT, Leverson GE, Heisey DM: Erythro-
poietin therapy may retard progression in chronic renal
transplant dysfunction. Nephrol Dial Transplant 17: 1667
1673, 2002
148. Choukroun G, Martinez F: Benefits of erythropoietin in
renal transplantation. Transplantation 79: S49S50, 2005
149. Muirhead N: Erythropoietin and renal transplantation.
Kidney Int Suppl 69: S86S92, 1999
150. Rodino MA, Shane E: Osteoporosis after organ transplan-
tation. Am J Med 104: 459469, 1998
151. K/DOQI clinical practice guidelines for bone metabolism
and disease in chronic kidney disease. Am J Kidney Dis 42:
S1S201, 2003
152. Maalouf NM, Shane E: Osteoporosis after solid organ
transplantation. J Clin Endocrinol Metab 90: 24562465, 2005
153. Palmer SC, Strippoli GF, McGregor DO: Interventions for
preventing bone disease in kidney transplant recipients: A
systematic review of randomized controlled trials. Am J
Kidney Dis 45: 638649, 2005
154. Cruz DN, Brickel HM, Wysolmerski JJ, Gundberg CG,
Simpson CA, Kliger AS, Lorber MI, Basadonna GP, Fried-
man AL, Insogna KL, Bia MJ: Treatment of osteoporosis
and osteopenia in long-term renal transplant patients with
alendronate. Am J Transplant 2: 6267, 2002
155. Jeffery JR, Leslie WD, Karpinski ME, Nickerson PW, Rush
DN: Prevalence and treatment of decreased bone density
in renal transplant recipients: A randomized prospective
trial of calcitriol versus alendronate. Transplantation 76:
14981502, 2003
156. Palmer S, McGregor DO, Strippoli GF: Interventions for
preventing bone disease in kidney transplant recipients.
Cochrane Database Syst Rev CD005015, 2005
157. Coco M, Glicklich D, Faugere MC, Burris L, Bognar I,
Durkin P, Tellis V, Greenstein S, Schechner R, Figueroa K,
McDonough P, Wang G, Malluche H: Prevention of bone
loss in renal transplant recipients: A prospective, random-
ized trial of intravenous pamidronate. J Am Soc Nephrol 14:
26692676, 2003
158. European best practice guidelines for renal transplanta-
tion. Section IV: Long-term management of the transplant
recipient. IV. 8. Bone disease. Nephrol Dial Transplant
17[Suppl 4]: 4348, 2002
159. Kilgo MS, Pirsch JD, Warner TF, Starling JR: Tertiary hy-
perparathyroidism after renal transplantation: Surgical
strategy. Surgery 124: 677683, 1998
160. Evenepoel P, Claes K, Kuypers D, Maes B, Vanrenterghem
Y: Impact of parathyroidectomy on renal graft function,
blood pressure and serum lipids in kidney transplant re-
cipients: A single centre study. Nephrol Dial Transplant 20:
17141720, 2005
161. Lee PP, Schiffmann L, Offermann G, Beige J: Effects of
parathyroidectomy on renal allograft survival. Kidney
Blood Press Res 27: 191196, 2004
162. Kruse AE, Eisenberger U, Frey FJ, Mohaupt MG: The cal-
cimimetic cinacalcet normalizes serum calcium in renal
transplant patients with persistent hyperparathyroidism.
Nephrol Dial Transplant 20: 13111314, 2005
163. Serra AL, Schwarz AA, Wick FH, Marti HP, Wuthrich RP:
Successful treatment of hypercalcemia with cinacalcet in
renal transplant recipients with persistent hyperparathy-
roidism. Nephrol Dial Transplant 20: 13151319, 2005
164. Green M, Avery R: Guidelines for the prevention and
management of infectious complications of solid organ
transplantation. Am J Transplant 4: S5S166, 2004
165. Fishman JA, Rubin RH: Infection in organ-transplant re-
cipients. N Engl J Med 338: 17411751, 1998
166. Shapiro R, Young JB, Milford EL, Trotter JF, Bustami RT,
Leichtman AB: Immunosuppression: Evolution in practice
and trends, 19932003. Am J Transplant 5: 874886, 2005
167. Kotton CN, Ryan ET, Fishman JA: Prevention of infection
in adult travelers after solid organ transplantation. Am J
Transplant 5: 814, 2005
168. Kasiske BL, Snyder JJ, Gilbertson DT, Wang C: Cancer after
kidney transplantation in the United States. Am J Trans-
plant 4: 905913, 2004
169. Morath C, Mueller M, Goldschmidt H, Schwenger V,
Opelz G, Zeier M: Malignancy in renal transplantation.
J Am Soc Nephrol 15: 15821588, 2004
170. Zeier M, Hartschuh W, Wiesel M, Lehnert T, Ritz E: Ma-
lignancy after renal transplantation. Am J Kidney Dis 39: E5,
2002
171. Danpanich E, Kasiske BL: Risk factors for cancer in renal
transplant recipients. Transplantation 68: 18591864, 1999
172. Dantal J, Soulillou JP: Immunosuppressive drugs and the
risk of cancer after organ transplantation. N Engl J Med 352:
13711373, 2005
173. Stallone G, Schena A, Infante B, Di Paolo S, Loverre A,
Maggio G, Ranieri E, Gesualdo L, Schena FP, Grandaliano
G: Sirolimus for Kaposis sarcoma in renal-transplant re-
cipients. N Engl J Med 352: 13171323, 2005
174. Mathew T, Kreis H, Friend P: Two-year incidence of ma-
Clin J Am Soc Nephrol 1: 623640, 2006 Medical Care of Kidney Transplant Recipients 639
lignancy in sirolimus-treated renal transplant recipients:
Results from five multicenter studies. Clin Transplant 18:
446449, 2004
175. Friedman AN, Miskulin DC, Rosenberg IH, Levey AS:
Demographics and trends in overweight and obesity in
patients at time of kidney transplantation. Am J Kidney Dis
41: 480487, 2003
176. Ducloux D, Kazory A, Simula-Faivre D, Chalopin JM: One-
year post-transplant weight gain is a risk factor for graft
loss. Am J Transplant 5: 29222928, 2005
177. Pirsch JD, Armbrust MJ, Knechtle SJ, DAlessandro AM,
Sollinger HW, Heisey DM, Belzer FO: Obesity as a risk
factor following renal transplantation. Transplantation 59:
631633, 1995
178. Meier-Kriesche HU, Arndorfer JA, Kaplan B: The impact of
body mass index on renal transplant outcomes: A signifi-
cant independent risk factor for graft failure and patient
death. Transplantation 73: 7074, 2002
179. European best practice guidelines for renal transplanta-
tion. Section IV: Long-term management of the transplant
recipient. IV. 5.7. Cardiovascular risks. Obesity and weight
gain. Nephrol Dial Transplant 17[Suppl 4]: 2930, 2002
180. El-Agroudy AE, Wafa EW, Gheith OE, Shehab el-Dein AB,
Ghoneim MA: Weight gain after renal transplantation is a
risk factor for patient and graft outcome. Transplantation
77: 13811385, 2004
181. Jindal RM, Zawada ET Jr: Obesity and kidney transplan-
tation. Am J Kidney Dis 43: 943952, 2004
182. Clunk JM, Lin CY, Curtis JJ: Variables affecting weight
gain in renal transplant recipients. Am J Kidney Dis 38:
349353, 2001
183. Alexander JW, Goodman HR, Gersin K, Cardi M, Austin J,
Goel S, Safdar S, Huang S, Woodle ES: Gastric bypass in
morbidly obese patients with chronic renal failure and
kidney transplant. Transplantation 78: 469474, 2004
184. Marterre WF, Hariharan S, First MR, Alexander JW: Gas-
tric bypass in morbidly obese kidney transplant recipients.
Clin Transplant 10: 414419, 1996
185. Lakka HM, Laaksonen DE, Lakka TA, Niskanen LK, Kum-
pusalo E, Tuomilehto J, Salonen JT: The metabolic syn-
drome and total and cardiovascular disease mortality in
middle-aged men. JAMA 288: 27092716, 2002
186. Ford ES, Giles WH, Dietz WH: Prevalence of the metabolic
syndrome among US adults: Findings from the third Na-
tional Health and Nutrition Examination Survey. JAMA
287: 356359, 2002
187. Third Report of the National Cholesterol Education Pro-
gram (NCEP) Expert Panel on Detection, Evaluation, and
Treatment of High Blood Cholesterol in Adults (Adult
Treatment Panel III) final report. Circulation 106: 3143
3421, 2002
188. Armstrong KA, Campbell SB, Hawley CM, Nicol DL, John-
son DW, Isbel NM: Obesity is associated with worsening
cardiovascular risk factor profiles and proteinuria progres-
sion in renal transplant recipients. Am J Transplant 5: 2710
2718, 2005
189. van Ree RM, de Vries AP, Oterdoom LH, The TH, Gan-
sevoort RT, Homan van der Heide JJ, van Son WJ, Ploeg RJ,
de Jong PE, Gans RO, Bakker SJ: Abdominal obesity and
smoking are important determinants of C-reactive protein
in renal transplant recipients. Nephrol Dial Transplant 20:
25242531, 2005
190. Keown P: Improving quality of life: The new target for
transplantation. Transplantation 72: S67S74, 2001
191. Valderrabano F, Jofre R, Lopez-Gomez JM: Quality of life
in end-stage renal disease patients. Am J Kidney Dis 38:
443464, 2001
192. Dew MA, Switzer GE, Goycoolea JM, Allen AS, DiMartini
A, Kormos RL, Griffith BP: Does transplantation produce
quality of life benefits? A quantitative analysis of the liter-
ature. Transplantation 64: 12611273, 1997
193. Matas AJ, Halbert RJ, Barr ML, Helderman JH, Hricik DE,
Pirsch JD, Schenkel FA, Siegal BR, Liu H, Ferguson RM:
Life satisfaction and adverse effects in renal transplant
recipients: A longitudinal analysis. Clin Transplant 16: 113
121, 2002
194. Hricik DE, Halbert RJ, Barr ML, Helderman JH, Matas AJ,
Pirsch JD, Schenkel FA, Siegal B, Ferguson RM: Life satis-
faction in renal transplant recipients: Preliminary results
from the transplant learning center. Am J Kidney Dis 38:
580587, 2001
195. Muehrer RJ, Becker BN: Life after transplantation: New
transitions in quality of life and psychological distress.
Semin Dial 18: 124131, 2005
196. Schweizer RT, Rovelli M, Palmeri D, Vossler E, Hull D,
Bartus S: Noncompliance in organ transplant recipients.
Transplantation 49: 374377, 1990
197. Butler JA, Roderick P, Mullee M, Mason JC, Peveler RC:
Frequency and impact of nonadherence to immunosup-
pressants after renal transplantation: A systematic review.
Transplantation 77: 769776, 2004
198. Chisholm MA, Vollenweider LJ, Mulloy LL, Jagadeesan M,
Wynn JJ, Rogers HE, Wade WE, DiPiro JT: Renal transplant
patient compliance with free immunosuppressive medica-
tions. Transplantation 70: 12401244, 2000
640 Clinical Journal of the American Society of Nephrology Clin J Am Soc Nephrol 1: 623640, 2006

Das könnte Ihnen auch gefallen