ArticlePDF Available

Higher tacrolimus concentrations early after transplant reduce the risk of acute GvHD in reduced-intensity allogeneic stem cell transplantation

Authors:

Abstract

There is significant variability in the serum concentrations of tacrolimus attained early post transplant due to drug interactions and genomic variation. We evaluated whether tacrolimus concentrations early post transplant correlated with incidence of acute GvHD in 120 consecutive patients allografted with a uniform reduced-intensity conditioning regimen. All patients received standard prophylaxis with oral tacrolimus and IV methotrexate. The primary variable of interest was mean weekly tacrolimus concentrations in the initial 4 weeks post transplant. In multivariate analysis, week 1 tacrolimus concentration was an independent predictor of acute grade 2–4 GvHD (hazard ratio (HR), 0.90; 95% confidence interval (CI), 0.84–0.97; P<0.01). This association was driven by a lower risk of acute grade 2–4 GvHD in patients with week 1 tacrolimus concentrations >12 ng/mL (HR, 0.47; 95% CI, 0.25–0.88; P=0.02). Week 1 tacrolimus concentrations were not associated with chronic GvHD, relapse or overall survival. Lower tacrolimus concentrations at weeks 2, 3 and 4 were not associated with a higher incidence of GvHD. In summary, we found that higher tacrolimus concentrations during the first week after allografting with a reduced-intensity conditioning regimen were associated with significantly reduced risk of acute grade 2–4 GvHD without increasing risk of relapse.
ORIGINAL ARTICLE
Higher tacrolimus concentrations early after transplant reduce
the risk of acute GvHD in reduced-intensity allogeneic stem
cell transplantation
A Ganetsky
1
, A Shah
1
, TA Miano
2
, W-T Hwang
3
,JHe
3
, AW Loren
4
, EO Hexner
4
, NV Frey
4
, DL Porter
4
and R Reshef
5
There is signicant variability in the serum concentrations of tacrolimus attained early post transplant due to drug interactions and
genomic variation. We evaluated whether tacrolimus concentrations early post transplant correlated with incidence of acute GvHD
in 120 consecutive patients allografted with a uniform reduced-intensity conditioning regimen. All patients received standard
prophylaxis with oral tacrolimus and IV methotrexate. The primary variable of interest was mean weekly tacrolimus concentrations
in the initial 4 weeks post transplant. In multivariate analysis, week 1 tacrolimus concentration was an independent predictor of
acute grade 24 GvHD (hazard ratio (HR), 0.90; 95% condence interval (CI), 0.840.97; Po0.01). This association was driven by a
lower risk of acute grade 24 GvHD in patients with week 1 tacrolimus concentrations 412 ng/mL (HR, 0.47; 95% CI, 0.250.88;
P= 0.02). Week 1 tacrolimus concentrations were not associated with chronic GvHD, relapse or overall survival. Lower tacrolimus
concentrations at weeks 2, 3 and 4 were not associated with a higher incidence of GvHD. In summary, we found that higher
tacrolimus concentrations during the rst week after allografting with a reduced-intensity conditioning regimen were associated
with signicantly reduced risk of acute grade 24 GvHD without increasing risk of relapse.
Bone Marrow Transplantation (2016) 51, 568572; doi:10.1038/bmt.2015.323; published online 21 December 2015
INTRODUCTION
The development of reduced-intensity conditioning (RIC) regi-
mens has led to the extended use of allogeneic hematopoietic
stem cell transplantation (HSCT), particularly in patients with
advanced age or those with signicant comorbidities. Although
RIC regimens are characterized by reduced toxicity, acute GvHD
remains a leading cause of morbidity and mortality in this type of
transplant.
1
Despite standard prophylactic measures, the rates of
acute GvHD are high, with incidence rates ranging from 25 to
68%.
24
Therefore, preventing GvHD without impairing the
graft-versus-tumor effect remains a critical goal for successful
HSCT.
Calcineurin inhibitors (CNIs) are considered to be the backbone
of GvHD prophylaxis in HSCT.
5
Successful administration of CNI is
complicated by their narrow therapeutic index and considerable
intra- and interpatient pharmacokinetic heterogeneity. The
unpredictable pharmacokinetic prole associated with CNI is the
result of drug interactions, genomic variation, hepatic and/or renal
function, and binding capacity to blood and plasma proteins.
69
This constellation of factors leads to signicant variability in CNI
concentrations attained within the rst week after HSCT, which
may affect outcomes as preclinical models have demonstrated
that the critical sequence of immunologic events that lead to
acute GvHD occurs within the rst few days after
transplantation.
10
Therefore, a delay in achieving therapeutic CNI
concentrations within the rst week post transplant may result in
a higher risk of acute GVHD.
Given these ndings, we hypothesized that CNI concentrations
attained early after transplant would affect clinical outcomes in
RIC HSCT recipients. Previous studies have shown conicting
results regarding the associations between CNI concentrations
and GVHD, possibly due to signicant variability in the studied
populations with respect to conditioning intensity, GVHD prophy-
laxis regimens, graft sources and CNI route of administration.
1114
To overcome some of these limitations and inform the
management of immunosuppression early after RIC HSCT, we
analyzed a uniform and large cohort of consecutive patients who
received oral tacrolimus (TAC) after peripheral blood stem cell
transplantation with udarabine+busulfan conditioning. The goal
of this analysis was to evaluate whether early TAC concentrations
correlated with incidence of GvHD, disease relapse and survival.
MATERIALS AND METHODS
Study population
We conducted a retrospective cohort study of 120 consecutive adult
patients undergoing rst allogeneic HSCT for a malignant hematologic
disorder at the University of Pennsylvania between January 2009 and
January 2014. All patients received a uniform RIC regimen of udarabine
(120 mg/m
2
) and busulfan (6.4 mg/kg), followed by infusion of T-cell
replete, G-CSF-mobilized peripheral blood stem cells from either a related
1
Department of Pharmacy, Hospital of the University of Pennsylvania, Philadelphia, PA, USA;
2
Center for Pharmacoepidemiology Research and Training, Perelman School of
Medicine, University of Pennsylvania, Philadelphia, PA, USA;
3
Department of Biostatistics and Epidemiology, Center for Clinical Epidemiology and Biostatistics, Perelman School of
Medicine, University of Pennsylvania, Philadelphia, PA, USA;
4
Blood and Marrow Transplantation Program, Abramson Cancer Center and the Division of Hematology and
Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA and
5
Blood and Marrow Transplantation Program and Columbia Center for
Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY, USA. Correspondence: Dr R Reshef, Blood and Marrow Transplantation
Program and Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, BB-1701C, 650 West 168th Street, New York,
NY 10032, USA.
Email: ran.reshef@columbia.edu
Received 1 September 2015; revised 12 November 2015; accepted 14 November 2015; published online 21 December 2015
Bone Marrow Transplantation (2016) 51, 568572
© 2016 Macmillan Publishers Limited All rights reserved 0268-3369/16
www.nature.com/bmt
or unrelated donor. All patients received standard GvHD prophylaxis with
oral TAC (0.06 mg/kg per day) in two divided doses starting 3 days before
HSCT and IV methotrexate at a dose of 15 mg/m
2
on day 1 and 10 mg/m
2
on days 3, 6 and 11. Antithymocyte globulin was not used in any of
the patients. The dose of TAC was adjusted to a target trough level of
515 ng/mL and was continued through 100 days post HSCT and then
tapered. The standard at our institution is to obtain the rst TAC trough
concentrations 1 day before HSCT and then at least twice weekly and at
least 72 h after any dosage change to allow drug levels to achieve
steady state. TAC whole-blood concentrations were measured by liquid
chromatographymass spectrometry. The study was approved by the
Institutional Review Board of the University of Pennsylvania.
Clinical outcomes
The primary outcome of interest was the correlation between mean
weekly TAC concentrations and acute grade 24 GvHD. We calculated
the mean TAC concentration for each of the rst 4 weeks post HSCT by
adding all of the available TAC concentrations from initiation of therapy
until the last day of the given week and dividing the sum by the total
number of available measurements. Secondary end points included acute
grade 34 GvHD, chronic GVHD, relapse, overall survival (OS), relapse-free
survival (RFS) and acute kidney injury (AKI). Acute GvHD was graded
according to the modied Glucksberg criteria and guidelines for data
collection recently published by the MAGIC Consortium.
15,16
Chronic GvHD
was graded according to the National Institutes of Health Consensus
Criteria.
17
Disease relapse was dened as morphological, cytogenetic
or radiological evidence of disease demonstrating pre-transplant
characteristics. We used the Disease Risk Index (DRI) stratication system
to classify patients according to disease type and disease status.
18
OS was
dened as the time interval between date of HSCT and death from any
cause or censored at last follow-up. RFS was dened as the time from date
of HSCT to death or relapse/progression, whichever came rst or censored
at last follow-up. AKI was dened as at least a twofold increase in serum
creatinine or a reduction in glomerular ltration rate by 450% from
baseline, as per the Risk, Injury, Failure, Loss and End-stage kidney disease
criteria.
19
Statistical analysis
Baseline and treatment characteristics were analyzed with descriptive
statistics. Associations between covariates and the cumulative incidence of
acute grade 24 GVHD were determined using Fine and Gray proportional
hazards regression. Death was considered a competing event. Associations
of covariates with OS and RFS were analyzed using Cox regression models.
Mean weekly TAC levels were included in the analyses as continuous
variables and then divided into tertiles. The following variables were
examined as potential covariates: patient and recipient age, patient and
recipient sex, disease type, donor source, DRI, degree of HLA match, and
presence or absence of CMV. Multivariable analysis was performed on
variables with a univariate P-valueo0.1. A two-sided P-value of 05 was
considered signicant for all other analyses. Competing risks regression
analyses were also conducted to identify predictors of relapse, allowing for
death as a competing event. Patients were censored at the time of donor
lymphocyte infusion for GvHD analyses. The associations between TAC
concentrations and AKI were conducted using t-tests.
RESULTS
Patient and transplant characteristics of the 120 subjects are
presented in Table 1. The median follow-up was 14.3 months
(range 0.766.3 months). The mean weekly TAC concentrations at
weeks 1, 2, 3 and 4 were 10.2 (range 2.819.4), 10.6 (range
3.923.9), 12.7 (range 4.224.1) and 11.9 (range 2.929.2) ng/mL,
respectively (Figure 1). Within this cohort, 115/120 (95.8%)
patients had complete TAC concentration data available for all
4 weeks of the analysis. In the remaining ve patients, the missing
TAC concentration values were conned to weeks 3 (n=1) and 4
(n= 4) of the study period. The majority of patients (111/120;
92.5%) received all four scheduled methotrexate doses.
GvHD
The primary outcome of interest was acute grade 24 GvHD.
The cumulative incidence of acute grade 24 GvHD was 21.3%
(95% condence interval (CI), 14.929.9%) at day 100 and 42.8%
(95% CI, 34.352.4%) at day 180 post HSCT. To assess whether
early TAC concentrations were predictive of acute grade 24
GvHD, we analyzed mean weekly TAC concentrations as
continuous variables up to 4 weeks post HSCT. We rst examined
the effect of TAC concentrations at each week independently, and
then constructed a multivariable model for predicting risk of acute
grade 24 GvHD. The hazard ratios (HRs) reect the increased or
decreased risk of acute grade 24 GvHD for each 1 ng/mL of
difference in mean TAC concentration.
In univariable analysis, week 1 TAC concentrations were
inversely associated with acute grade 24 GvHD (HR, 0.92; 95%
CI, 0.850.99; P= 0.03). Other variables associated with a higher
risk of acute grade 24 GvHD that met our threshold for modeling
were the presence of a single-allele HLA mismatch (HR, 2.14; 95%
CI, 1.203.84; Po0.01) and lymphoid malignancies as opposed to
Table 1. Patient and transplant characteristics (N=120)
Characteristic Value
Recipient age, median in years (range) 62 (2872)
Recipient sex, male/female (n) 70/50
Donor age, median in years (range) 44 (1872)
Donor sex, male/female (n) 64/56
Sex mismatch, n(%) 55 (46)
Female donor to male recipient transplant, n(%) 28 (24)
Diagnosis, n(%)
Acute myeloid leukemia 49 (41)
Myelodysplastic syndromes 33 (27)
Non-Hodgkin lymphoma 22 (18)
Acute lymphoblastic leukemia 8 (7)
Other
a
8 (7)
Donor source, n(%)
Matched sibling 53 (44)
Matched unrelated 50 (42)
Single-allele mismatched unrelated 17 (14)
Disease Risk Index, n(%)
Low 10 (8)
Intermediate 80 (67)
High/very high 30 (25)
a
Other includes: chronic myeloid leukemia (n=2), Hodgkin lymphoma
(n=2), multiple myeloma (n=1), myelodysplastic/myeloproliferative neo-
plasm (n=1) and myelobrosis (n=2).
30
25
20
15
10
5
0
Week 1 Week 2 Week 3 Week 4
Week after transplant
Mean tacrolimus concentration (ng/mL)
N=120 N=120 N=119 N=116
Figure 1. Signicant variability in TAC concentrations attained early
after RIC HSCT. Box-and-whisker plot showing the distribution of
mean weekly TAC concentrations during the rst 4 weeks after
transplantation.
Tacrolimus and acute GvHD
A Ganetsky et al
569
© 2016 Macmillan Publishers Limited Bone Marrow Transplantation (2016) 568 572
myeloid malignancies (HR, 0.56; 95% CI, 0.301.03; P= 0.06). When
incorporating these covariates into a multivariable model, higher
week 1 TAC concentrations remained independently associated
with a lower risk of acute grade 24 GvHD (HR, 0.90; 95% CI,
0.840.97; Po0.01), as shown in Figure 2. We found no
correlations between lower TAC concentrations at weeks 2, 3 or
4 and increased incidence of acute grade 24 GvHD. In addition,
TAC concentrations on the day before HSCT were not associated
with acute grade 24 GVHD.
To further characterize the relationship between week 1 TAC
concentrations and acute grade 24 GVHD, we examined the
effect of week 1 TAC concentrations categorized in tertiles ( o8.5,
8.512 and 412 ng/mL). Interestingly, the inverse association
between week 1 TAC concentrations and acute grade 24 GVHD
was driven by a lower risk in the upper tertile (412 ng/mL), as
shown in Figure 3. Patients in the upper tertile had a lower risk of
acute grade 24 GVHD compared with those in the lower tertile
(HR, 0.47; 95% CI, 0.250.88; P= 0.02). There was no difference in
risk of acute grade 24 GVHD when comparing the intermediate
and lower tertiles (HR, 1.1; 95% CI, 0.601.93; P= 0.80).
We then analyzed the effect of week 1 TAC concentrations on
acute grade 24 GvHD in recipients of grafts from related and
unrelated donors separately. The cumulative incidence of acute
grade 24 in related donor HSCT at day 100 and day 180 was
11.3% (95% CI, 2.520.1%) and 32.1% (19.145.1%), respectively.
Week 1 TAC concentrations were not predictive of acute grade
24 GVHD following related donor HSCT (adjusted HR, 0.96; 95%
CI, 0.851.07; P= 0.46). In recipients of grafts from unrelated
donors, the cumulative incidence of acute grade 24 GVHD at day
100 was 31.3% (95% CI, 19.942.7%) and at day 180 was 50.7%
(38.363.1%). In unrelated donor HSCT, higher week 1 TAC
concentrations were associated with a lower risk of acute grade
24 GvHD (adjusted HR, 0.88; 95% CI, 0.810.96; P= 0.003).
Furthermore, we evaluated whether TAC concentrations
within the rst month post HSCT impacted the risk of acute
grade 34 GVHD. The cumulative incidence of acute grade 34
GVHD was 6.8% (95% CI, 3.210.4%) at day 100 and 16.6% (95% CI,
10.223.0%) at day 180 post HSCT. We found no association
between TAC concentrations and this outcome (data not shown).
We then examined the associations between TAC concentrations
and acute grade 34 GVHD when recipients of related and
unrelated grafts were analyzed separately. In related donor HSCT,
the cumulative incidence of acute grade 34 at day 100 was 5.8%
(95% CI, 0.611.0%) and 15.2% (5.824.6%) day 180. The
cumulative incidence of acute grade 34 GvHD in recipients of
grafts from unrelated donors was 7.9% (2.912.9%) at day 100 and
18.9% (10.127.7%) at day 180. Week 1 TAC concentrations were
not predictive of acute grade 34 GVHD when patients were
analyzed separately according to donor source. Similar analyses
were conducted for chronic GvHD and no associations were found
(data not shown).
Relapse
Because the pathogenesis of GvHD is closely intertwined with
the graft-versus-tumor effect, we examined whether early TAC
concentrations after RIC HSCT inuenced risk of disease relapse.
The cumulative incidence of disease relapse was 29.2% (95% CI,
21.837.9%) at day 180 and 38.3% (95% CI, 30.147.3%) at 1 year.
In univariable analyses, disease relapse was not associated with
mean TAC concentrations at week 1 (HR, 0.98; 95% CI, 0.891.06;
P= 0.58), week 2 (HR, 0.98; 95% CI, 0.911.05; P= 0.52), week 3
(HR, 0.97; 95% CI, 0.911.03; P= 0.29) or week 4 (HR, 1.04; 95% CI,
0.981.11; P= 0.15). We also analyzed the relationship between
TAC concentrations and risk of relapse with adjustment for the
DRI and found no signicant associations (data not shown).
In addition, mean TAC concentrations did not correlate with
donorrecipient whole-blood or T-cell chimerism levels at days 30,
60 and 100, and 1 year after RIC HSCT.
RFS and OS
The 2-year estimated rates of RFS and OS were 25.8% (95% CI,
18.834.4%) and 34.2% (95% CI, 26.343.0%), respectively. In
univariable analysis, higher week 3 TAC concentrations were
associated with improved RFS (HR, 0.95; 95% CI, 0.901.00;
P= 0.06) but did not meet our threshold for statistical signicance.
With adjustment for the DRI, this correlation remained
non-signicant (HR, 0.95; 95% CI, 0.901.01; P= 0.08). TAC
concentrations at weeks 1, 2 and 4 were not predictive of RFS.
We conducted a similar analysis to identify associations
between early TAC concentrations and OS. In univariable analysis,
higher week 3 TAC concentrations were associated with improved
OS (HR, 0.95; 95% CI, 0.891.00; P= 0.07) but did not reach
statistical signicance. With adjustment for disease type, recipient
age and DRI, the relationship between higher week 3 TAC
concentrations and improved OS remained non-signicant
(HR, 0.94; 95% CI, 0.891.00; P= 0.06). TAC concentrations at
weeks 1, 2 and 4 were not associated with OS.
Week 1 TAC
Week 2 TAC
Week 3 TAC
Week 4 TAC
0.5 1 2
Adjusted hazard ratio
P<0.01
P=0.16
P=0.66
P=0.52
Figure 2. Signicant association between week 1 TAC concentra-
tions and acute grade 24 GvHD. Multivariable analysis showing
adjusted hazard ratios (aHRs) for acute grade 24 GvHD based on
mean TAC concentrations at weeks 1, 2, 3 and 4 after RIC HSCT. The
aHRs reect the increased or decreased risk of acute grade 24
GVHD for each 1 ng/mL of difference in mean TAC concentration.
Mean week 1 TAC levels
<8.5 ng/mL (low)
8.5-12 ng/mL (intermediate)
>12 ng/mL (high)
1.0
0.8
0.6
0.4
0.2
0.0
0123456
Cumulative incidence
Time from transplant (months)
Figure 3. Lower risk of acute grade 24 GvHD in patients with
mean week 1 TAC412 ng/mL. Cumulative incidence plots showing
acute grade 24 GvHD according to mean week 1 TAC concentra-
tions. Patients in the lower tertile ( o8.5 ng/mL), middle tertile
(8.512 ng/mL) and upper tertile (412 ng/mL) are represented by
the dashed, dotted and solid lines, respectively.
Tacrolimus and acute GvHD
A Ganetsky et al
570
Bone Marrow Transplantation (2016) 568 572 © 2016 Macmillan Publishers Limited
Kidney injury
As nephrotoxicity is a commonly reported complication of TAC,
we examined whether TAC concentrations early post transplant
were predictive of AKI in this cohort. Renal impairment, dened by
the Risk, Injury, Failure, Loss and End-stage kidney disease criteria,
which is a standard criterion in pharmacological studies, occurred
in 20 (16.7%) patients during the rst 4 weeks after RIC HSCT.
Week 1 TAC concentrations were higher in patients who
developed AKI by week 2 post transplant compared with those
who did not (12.7 vs 10.0 ng/mL; P= 0.01). We did not observe an
association between TAC concentrations at any other time points
and AKI within the rst month post transplant. No patients
required hemodialysis within the rst 4 weeks after HSCT. We also
examined whether TAC concentrations early after RIC HSCT were
associated with chronic renal impairment. Of the 71 patients alive
at 1 year after HSCT, no one had a serum creatinine 42 mg/dL.
DISCUSSION
In this study, we demonstrated that there is signicant variability
in the serum concentrations of TAC attained early post-RIC
HSCT. In addition, we found that higher TAC concentrations
during the rst week after RIC HSCT were associated with
signicantly reduced risk of acute grade 24 GvHD without
increasing risk of relapse. For each 1 ng/mL increase in TAC
concentration, there was a 10% decrease in risk of acute grade 24
GvHD. Importantly, this association was driven by a lower risk of
acute grade 24 GvHD in patients with mean week 1 TAC
concentrations 412 ng/mL. To the best of our knowledge, this
study is the rst to characterize the importance of achieving
higher therapeutic TAC concentrations within the rst week after
RIC HSCT.
As expected, we observed a direct association between week 1
TAC concentrations and incidence of AKI occurring by day 14 post
transplant. The incidence of AKI has been previously reported to
be proportionally related to TAC concentrations in HSCT
recipients.
20
AKI was reversible and long-term renal complications
were not observed.
The results of our study show the critical importance of
achieving therapeutic TAC concentrations within the rst week
after RIC HSCT to optimally attenuate donor alloreactivity.
This association is consistent with preclinical studies that
demonstrated that the initiating events of acute GvHD occur very
early after transplant.
10,21
It was observed that alloreactive T-cell
activation, proliferation and migration to GvHD target organs
occur within several days after stem cell infusion. In addition,
ex vivo analyses of gastrointestinal tract tissue have shown
alloreactive CD4+ donor lymphocyte inltration of Peyers patches
and mesenteric lymph nodes as early as 12 h after HSCT.
10
The
importance of inhibiting alloreactivity within the rst week after
RIC HSCT has been further characterized with the emergence of
novel approaches to GvHD prophylaxis, including proteasome
inhibition with bortezomib and post-transplant cyclophospha-
mide. The efcacy of both of these novel strategies is critically
dependent on the early timing of drug administration after
HSCT.
22,23
Taken together, our ndings imply that achieving a
mean TAC concentration 412 ng/mL during the rst week after
RIC HSCT potentially mitigates the intense alloreactivity that
occurs immediately after stem cell infusion and thereby reduces
the risk of developing acute GvHD. In a subset analysis, this
association appeared to be driven by the group of recipients of
unrelated donor grafts, although the analysis in recipients of
related grafts may have been inadequately powered due to a
smaller sample size.
The optimal target concentration of CNI early after HSCT
has been previously examined with conicting results.
1114
The varying results are likely due to heterogeneity in studied
populations, inclusion of multiple GvHD prophylaxis regimens,
different routes of TAC administration and heterogeneity in
conditioning regimens and graft sources. To overcome these
limitations, our analysis focused on a homogeneous patient
population consisting of patients undergoing rst allogeneic
HSCT who were allografted with udarabine/busulfan, the most
commonly used RIC regimen according to the Center for
International Blood and Marrow Transplant Research.
1
In addition,
all patients received a uniform GVHD prophylaxis regimen and
received peripheral blood stem cells. Another retrospective study
that reported on a uniform patient population that underwent RIC
HSCT found no associations between TAC concentrations and
acute grade 24 GvHD, although week 2 TAC concentrations
o10.5 ng/mL were associated with a higher risk of acute grade
34 GvHD. In contrast to our ndings, there was no correlation
between week 1 TAC concentrations and acute GvHD.
11
Several
differences between this report and our study, including the use
of non-myeloablative conditioning (low-dose TBI ± udarabine)
and the use of mycophenolate mofetil and not methotrexate,
could potentially explain the different results of these studies.
Our data highlight the importance of developing novel
methods to optimize the initial dosing of TAC in RIC HSCT
recipients. One approach that may aid in achieving higher
therapeutic concentrations rapidly is to incorporate a patients
genotype into the formula for determining the initial starting dose
rather than using the standard weight-based xed-dose strategy.
TAC is primarily metabolized by cytochrome P450 3A4 and 3A5,
both highly polymorphic isoenzymes. In addition, a number
of other enzymes responsible for TAC metabolism possess
gene variants that have the propensity to inuence TAC
pharmacokinetic.
24
Identifying patient-specic genotypes prior
to initiating TAC may reduce the time it takes to achieve
therapeutic concentrations of TAC and thereby reduce the
incidence of acute GvHD.
In summary, we conclude that achieving higher TAC con-
centrations, and in particular levels 412 ng/mL within the rst
week of RIC HSCT, may signicantly reduce the risk of acute grade
24 GvHD without impairing the graft-versus-tumor effect.
Although post-transplant AKI is more commonly seen in patients
with high TAC concentrations, long-term renal complications are
rare. These data highlight the importance of optimizing the initial
dosing of TAC in RIC HSCT recipients. Prospective conrmation of
our ndings is warranted.
CONFLICT OF INTEREST
The authors declare no conict of interest.
ACKNOWLEDGEMENTS
This work was supported by a Career Development Award from the Conquer Cancer
Foundation (RR); Amy Strelzer Manasevit Award from the National Marrow Donor
Program (RR); National Institutes of Health grants K23-CA178202 (RR) &
U01-HL069286 (DLP), and the Margie and Andy Rooke Fund for Leukemia Research
(RR and DLP). We thank Oren Litvin for help with preparation of the gures.
REFERENCES
1 Reshef R, Porter DL. Reduced-intensity conditioned allogeneic SCT in adults
with AML. Bone Marrow Transplant 2015; 50: 759769.
2 Jagasia M, Arora M, Flowers EM, Chao NJ, McCarthy PL, Cutler CS et al. Risk factors
for acute GVHD and survival after hematopoietic cell transplantation. Blood 2012;
119:296307.
3 Eapen M, Logan BR, Horowitz MM, Zhong X, Perales MA, Lee SJ et al. Bone marrow
or peripheral blood for reduced-intensity conditioning unrelated donor
transplantation. J Clin Oncol 2015; 33:364369.
4 Johnston L. Acute graft-versus-host disease: differing risk with differing graft
sources and conditioning intensity. Best Pract Res Clin Haematol 2008; 21:
177192.
Tacrolimus and acute GvHD
A Ganetsky et al
571
© 2016 Macmillan Publishers Limited Bone Marrow Transplantation (2016) 568 572
5 Choi SW, Reddy P. Current and emerging strategies for the prevention of
graft-versus host disease. Nat Rev Clin Oncol 2014; 11: 536547.
6 Jacobson P, Ng J, Ratanatharathorn V, Uberti J, Brundage RC. Factors affecting the
pharmacokinetics of tacrolimus (FK506) in hematopoietic cell transplant (HCT)
patients. Bone Marrow Transplant 2001; 28:753758.
7 Onizuka M, Kunii N, Toyosaki M, Machida S, Ohgiya D, Ogawa Y et al. Cytochrome
P450 genetic polymorphisms inuence the serum concentration of calcineurin
inhibitors in allogeneic hematopoietic SCT recipients. Bone Marrow Transplant
2011; 46: 11131117.
8 Brown NW, Gonde CE, Adams JE, Tredger JM. Low hematocrit and serum albumin
concentrations underlie the overestimation of tacrolimus concentrations by
microparticle enzyme immunoassay versus liquid chromatography-tandem mass
spectrometry. Clin Chem 2005; 51: 586592.
9 Miano TA, Ganetsky A, Porter DL, Reshef R. Serum hemoglobin is a predictor of
tacrolimus whole blood concentration in hematopoietic stem cell transplant
patients. 2015 American Society for Clinical Pharmacology and Therapeutics Annual
Meeting. 1 February 2015; Wiley-Blackwell. 2015, pp S80S80.
10 Beilhack A, Schulz S, Baker J, Beilhack GF, Wieland CB, Herman EI et al. In vivo
analyses of early events in acute graft-versus-host disease reveal sequential
inltration of T-cell subsets. Blood 2005; 106:11131122.
11 Ram R, Storer B, Mielcarek M, Sandmaier BM, Maloney DG, Martin PJ et al.
Association between calcineurin inhibitor blood concentrations and outcomes
after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant
2012; 18: 414422.
12 Offer K, Kolb M, Jin Z, Bhatia M, Kung AL, George D et al. Efcacy of tacrolimus/
mycophenolate mofetil as acute graft-versus-host disease prophylaxis and the
impact of subtherapeutic tacrolimus levels in children after matched sibling
donor allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant
2015; 21: 496502.
13 Mori T, Kato J, Shimizu T, Aisa Y, Nakazato T, Yamane A et al. Effect of
early posttransplantation tacrolimus concentration on the development of
acute graft-versus-host disease after allogeneic hematopoietic stem cell
transplantation from unrelated donors. Biol Blood Marrow Transplant 2012; 18:
229234.
14 Rogosheske JR, Fargen AD, DeFor TE, Warlick E, Arora M, Blazar BR et al.
Higher therapeutic CsA levels early post-transplantation reduces risks of acute
graft-versus-host disease and improves survival. Bone Marrow Transplant 2014; 49:
122125.
15 Przepiorka D, Weisdorf D, Martin P, Klingemann HG, Beatty P, Hows J et al. 1994
Consensus conference on acute GVHD grading. Bone Marrow Transplant 1995; 15:
825828.
16 Harris AC, Young R, Devine S, Hogan WJ, Ayuk F, Bunworasate U et al.
International, multi-center standardization of acute graft-versus-host disease
clinical data collection: a report from the MAGIC consortium. Biol Blood Marrow
Transplant (e-pub ahead of print 16 September 2015; doi:10.1016/j.
bbmt.2015.09.001).
17 Filipovich AH, Weisdorf D, Pavletic S, Socie G, Wingard JR, Lee SJ et al. National
Institutes of Health consensus development project on criteria for clinical trials in
chronic graft-versus-host disease: I. Diagnosis and staging working group report.
Biol Blood Marrow Transplant 2005; 11:945956.
18 Armand P, Kim HT, Logan BR, Wang Z, Alyea EP, Kalaycio ME et al. Validation and
renement of the disease risk index for allogeneic stem cell transplant ation. Blood
2014; 123:36643671.
19 Bellomo R, Ronco C, Kellum JA, Mehta RL, Palevsky P. Acute renal failure -
denition, outcome measures, animal models, uid therapy and information
technology needs: the Second International Consensus Conference of the Acute
Dialysis Quality Initiative (ADQI) Group. Crit Care 2004; 8: R204R212.
20 Przepiorka D, Nash RA, Wingard JR, Zhu J, Maher RM, Fitzsimmons WE et al.
Relationship of tacrolimus whole blood levels to efcacy and safety outcomes
after unrelated donor marrow transplantation. Biol Blood Marrow Transplant 1999;
5:9497.
21 Murai M, Yoneyama H, Ezaki T, Suematsu M, Terashima Y, Harada A et al. Peyers
patch is the essential site in initiating murine acute and lethal graft-versus-host
reaction. Nat Immunol 2003; 4: 154160.
22 Sun K, Wilkins DEC, Anver MR, Sayers TJ, Panoskaltsis-Mortari A, Blazar BR et al.
Differential effects of proteasome inhibition by bortezomib on murine acute
graft-versus-host disease (GVHD): delayed administration of bortezomib results in
increased GVHD-dependent gastrointestinal toxicity. Blood 2005; 106: 32933299.
23 Luznik L, ODonnell PV, Fuchs EJ. Post-transplantation cyclophosphamide for
tolerance induction in HLA-haploidentical BMT. Semin Oncol 2012; 39:116.
24 Hesselink DA, Bouamar R, Elens L, van Schaik RH, van Gelder T. The role of
pharmacogenetics in the disposition of and response of tacrolimus in solid organ
transplantation. Clin Pharmacokinet 2014; 53:123139.
Tacrolimus and acute GvHD
A Ganetsky et al
572
Bone Marrow Transplantation (2016) 568 572 © 2016 Macmillan Publishers Limited
... Less is known, however, regarding its optimal C 0 when Tac is combined with PTCy. Moreover, although higher maintained Tac blood concentrations have been associated with Tac toxicity and transplant-related mortality (TRM), no consensus exists regarding the impact of low Tac concentrations on the incidence of GVHD [11,12,[15][16][17]. ...
... Studies have evaluated the optimal range of blood concentrations of Tac early after alloHSCT for preventing GVHD. Although several of these studies reported that the incidence of aGVHD was unaffected by Tac concentrations, more recent studies have shown an association between low Tac levels and a higher risk of aGVHD [15][16][17]25,26]. For example, an analysis of a group of patients who underwent HLA-matched unrelated donor transplantation, most of whom were administered a MAC regimen and all of whom received intravenous Tac and short-term MTX, with a target Tac level set at 10 to 20 ng/mL, found that low mean Tac blood concentrations during the second and third weeks after transplantation were significant risk factors for aGVHD [16]. ...
... For example, an analysis of a group of patients who underwent HLA-matched unrelated donor transplantation, most of whom were administered a MAC regimen and all of whom received intravenous Tac and short-term MTX, with a target Tac level set at 10 to 20 ng/mL, found that low mean Tac blood concentrations during the second and third weeks after transplantation were significant risk factors for aGVHD [16]. In patients undergoing RIC alloHSCT using Tac plus MTX, achievements of Tac concentrations >12 ng/mL within the first week after transplant were found to significantly reduce the risk of acute grades II-IV GVHD without impairing the graft-versus-tumor effect [15]. In the latter study, however, lower Tac concentrations at weeks 2, 3, and 4 were not associated with higher rates of GVHD. ...
Article
Full-text available
Tacrolimus (Tac) is a pivotal immunosuppressant agent used to prevent graft-versus-host disease (GVHD) after allogeneic stem cell transplantation (alloHSCT). Tac is characterized by a narrow therapeutic window and a high inter-patient and intra-patient pharmacokinetic variability (IPV). Although high IPV of Tac concentrations has been associated with adverse post-transplant outcomes following solid organ transplantation, the effects of Tac IPV on alloHSCT recipients have not been determined. Tac IPV was therefore retrospectively evaluated in 128 alloHSCT recipients receiving high-dose post-transplant cyclophosphamide (PTCy) and the effects of Tac IPV on the occurrence of acute GVHD (aGVHD) were analyzed. Tac IPV was calculated from pre-dose concentrations (C0) measured during the first month after Tac initiation. The cumulative rates of grades II-IV and grades III-IV aGVHD at day +100 were 22.7% and 7%, respectively. Higher Tac IPV was associated with a greater risk of developing GVHD, with patients having IPV > 50th percentile having significantly higher rates of grades II-IV (34.9% vs. 10.8%; hazard ratio [HR] 3.858, p < 0.001) and grades III-IV (12.7% vs. 1.5%; HR 9.69, p = 0.033) aGVHD than patients having IPV ≤ 50th percentile. Similarly, patients with IPV > 75th percentile had higher rates of grades II-IV (41.9% vs. 16.5%; HR 3.30, p < 0.001) and grades III-IV (16.1% vs. 4.1%; HR 4.99, p = 0.012) aGVHD than patients with IPV ≤ 75th percentile. Multivariate analyses showed that high Tac IPV (>50th percentile) was an independent risk factor for grades II-IV (HR 2.99, p = 0.018) and grades III-IV (HR 9.12, p = 0.047) aGVHD. Determination of Tac IPV soon after alloHSCT could be useful in identifying patients at greater risk of aGVHD.
... Despite the increased use of PTCy combined with TAC, there are currently no standard dosing guidelines or therapeutic target serum levels for TAC. It is assumed that immunosuppressive levels pre-engraftment could impact T cell repertoire and recovery, which may dictate the incidence and severity of GvHD [22][23][24][25][26][27][28][29], the graft-versus-leukemia (GvL) effect and transplant outcomes. TISS represents an early adjustable timepoint where intervention could lead to better outcomes. ...
... Thus, it is beneficial to determine optimal TISS. When combined with methotrexate, higher early (first week) TAC level was associated with lower risk of aGvHD grade II-IV in RIC setting [22,29], but less GvL with higher RR [29]. We observed a trend of better DFS with lower RR in patients with TISS < 10 ng/ mL vs. ≥10 ng/mL without affecting GvHD and NRM. ...
... One reasonable explanation is that early lower levels of suppression (TISS < 10 ug/mL) permitted GvL that likely contributed to lower RR. TISS was not correlated with aGvHD or cGvHD in our analysis, as noted in TAC/methotrexate-based regimen [22,29], which is possibly due to the upfront effect of PTCy on T cells before TAC is introduced. In our analysis, PBSCs as the graft source was the only predictor of risk of aGvHD grade II-IV, which is comparable to published data [5,7]. ...
Article
Full-text available
Post-transplant cyclophosphamide (PTCy) combined with tacrolimus (TAC) as graft-versus-host disease (GvHD) prophylaxis post-hematopoietic cell transplantation (HCT) is safe and effective. Optimal serum levels of TAC in this combination remain undetermined. We hypothesized that TAC at initial steady state (TISS) of <10 ng/mL could promote optimal transplant outcomes and prevent TAC-associated toxicities. We retrospectively analyzed a consecutive case series of 210 patients who received PTCy/TAC-based prophylaxis post-HCT from 1/2013-6/2018. Patients received HCT from haploidentical (n = 172) or mismatched donors (n = 38), and flat dose (FD) or weight-based dose (WBD) TAC. Twenty-four-month overall survival (OS), disease free survival (DFS), and relapse rate (RR) were 61%, 56%, and 22%, respectively, in TISS < 10 ng/mL cohort (n = 176), and 50%, 43%, and 35%, respectively, in TISS ≥ 10 ng/mL cohort (n = 34) (OS, P = 0.71; DFS, P = 0.097; RR, P = 0.031). OS, DFS, RR, non-relapse mortality, acute GvHD grade II–IV, grade III–IV or chronic GvHD by TISS were similar in multivariable analysis. TISS ≥ 10 ng/mL conferred increased risk of viral infection (P = 0.003). More patients receiving FD vs. WBD had TISS < 10 ng/mL (P = 0.001). Overall, TISS < 10 ng/mL early post HCT conferred similar survival outcomes and lowered risk of viral infection and toxicities compared to TISS ≥ 10 ng/mL.
... A study in the adult population reported that the early post-transplantation blood concentrations of TAC had a significant impact on the development of aGVHD [12], with another reporting that a level of <5 ng/mL was associated with increased aGVHD [13]. In an attempt to understand the impact of early (first four weeks) TAC levels on aGVHD incidence, Ganetsky et al. showed in 120 patients undergoing allo HSCT that had a TAC concentration of ≥12 ng/mL during the first week post-transplantation was associated with reduced risk of aGVHD [14]. However, Mori et al. showed that in 60 patients undergoing allo-HSCT, the mean blood concentration of TAC (17.3 ± 2.1) during the third week after allo-HSCT was significantly associated with lower aGVHD [12]. ...
... Specifically, TAC levels of ≥10.15 ng/mL were associated with a lower risk of grade II-IV and III-IV aGVHD. In contrast to the findings of others [14,25,26], we did observe TAC levels ≥10.15 ng/mL were also marginally associated with a lower incidence of extensive cGVHD. Non-myeloablative conditioning appears to have minimum tissue injury [27], a transient state of mixed chimerism [28], a different prophylaxis regimen [29], which could, in turn, lead to delayed GVHD. ...
... Similar results were reported by Ganetsky et al., where they showed that TAC concentrations >12 ng/mL during the first week after allotransplantation was associated with significantly reduced risk of II-IV aGVHD without increasing risk of relapse. In contrast to our study, Ganetsky et al. [14] did not find any association between TAC and cGVHD or relapse. Another difference was that all patients received reduced-intensity conditioning, whereas, in our study, there was a mix of patients receiving myeloablative or reduced-intensity conditioning. ...
Article
Full-text available
Simple Summary Allogeneic hematopoietic stem cell transplantation is a potentially curative treatment for many hematological malignancies and disorders but is often complicated by a relapse of the underlying disease, graft-vs-host disease and infectious complications. However, despite the introduction of calcineurin inhibitors such as tacrolimus, graft-versus-host disease remains one of the major life-threatening complications of allogeneic hematopoietic stem cell transplantation. Due to a variety of factors, there is variability in tacrolimus concentrations during the early weeks post-transplantation. Since the immunologic events leading to acute GVHD also occur in the first few days post-transplantation, it is important that optimal levels be attained early after transplantation. The findings from this study will help inform the management of optimal tacrolimus levels to be attained early post-transplantation. Abstract Acute graft versus host disease (aGVHD) remains a leading cause of morbidity and mortality in allogeneic hematopoietic stem cell transplant (allo-HSCT). Tacrolimus (TAC), a calcineurin inhibitor that prevents T-cell activation, is commonly used as a GVHD prophylaxis. However, there is variability in the serum concentrations of TAC, and little is known on the impact of early TAC levels on aGVHD. We retrospectively analyzed 673 consecutive patients undergoing allo-HSCT at the Ohio State University between 2002 and 2016. Week 1 TAC was associated with a lower risk of aGVHD II–IV at TAC level ≥10.15 ng/mL (p = 0.03) compared to the lowest quartile. The cumulative incidence of relapse at 1, 3 and 5 years was 33%, 38% and 41%, respectively. TAC levels at week 2, ≥11.55 ng/mL, were associated with an increased risk of relapse (p = 0.01) compared to the lowest quartile. Subset analysis with acute myeloid leukemia and myelodysplastic syndrome patients showed significantly reduced aGVHD with TAC level ≥10.15 ng/mL at week 1 and a higher risk of relapse associated with week 2 TAC level ≥11.55 ng/mL (p = 0.02). Hence, achieving ≥10 ng/mL during the first week of HCT may mitigate the risk of aGVHD. However, levels (>11 ng/mL) beyond the first week may be associated with suppressed graft versus tumor effect and higher relapse.
... CSA and TAC are considered equivalent and there has been discussions on the target serum concentrations. It has been shown that the target concentrations of TAC in the first 4 weeks post-ASCT were associated with GVHD [48,61]. For CSP-A, concentrations of 200-300 μg/L are usually recommended when the total daily dose is divided into 2 doses 12 hours apart f for preventing aGVHD [43]. ...
... A study in the adult population reported that the early post-transplantation blood concentrations of TAC had a significant impact on the development of aGVHD with another reporting TAC level of <5ng/ml was associated with increased acute GVHD [68,69]. In an attempt to understand the impact of early TAC levels on aGVHD incidence, Ganetsky et al. [61] showed that TAC concentration ≥12 ng/ml during the first week post-transplantation was associated with reduced risk of aGVHD. In our institutional study with approx. ...
Article
Full-text available
Graft-versus host disease is one of the major causes of death in patients undergoing allogeneic hematopoietic stem cell transplantation. Major contributing factors to severity of graft-versus host disease include HLA mismatch, conditioning regimen intensity, age of donor and recipient, source of stem cell, and donor type. Strategies and clinical trials are ongoing to reduce the incidence of graft-versus host disease, and to improve on the survival outcome of patients with graft-versus host disease, Research efforts continue to develop new ways of identification, prevention, and treatment for graft versus host disease. In addition, efforts are also being made to incorporate biomarker for the early detection of graft versus host disease. In addition, strategies utilizing monoclonal antibodies and cytokines are being tested as potential therapeutic options. Here we provide evidence for recommended regimens for graft versus host disease, prophylaxis and treatment.
... Unlike in solid organ transplantation (SOT), in alloHSCT Tac is maintained for the first 3-6 months post-transplant and then tapered and discontinued in the absence of GVHD by the 6th-12th month post-transplant [6]. Therefore, exposure to Tac during the immediate period after the HSCT period is of utmost importance to prevent subsequent GVHD [4,7,8]. Furthermore, subtherapeutic Tac trough concentrations (C 0 < 5 ng/mL) at 48 h or 7 days after transplant has been associated with an increased incidence of acute GVHD (GVHD) [9,10]. ...
Article
Full-text available
Tacrolimus (Tac) is pivotal in preventing acute graft-versus-host disease (GVHD) after allogeneic hematopoietic stem cell transplantation (alloHSCT). It has been reported that genetic factors, including CYP3A5*3 and CYP3A4*22 polymorphisms, have an impact on Tac metabolism, dose requirement, and response to Tac. There is limited information regarding this topic in alloHSCT. The CYP3A5 genotype and a low Tac trough concentration/dose ratio (Tac C0/D ratio) can be used to identify fast metabolizers and predict the required Tac dose to achieve target concentrations earlier. We examined 62 Caucasian alloHSCT recipients with a fast metabolizer phenotype (C0/dose ratio ≤ 1.5 ng/mL/mg), assessing CYP3A5 genotypes and acute GVHD incidence. Forty-nine patients (79%) were poor metabolizers (2 copies of the variant *3 allele) and 13 (21%) were CYP3A5 expressers (CYP3A5*1/*1 or CYP3A5*1/*3 genotypes). CYP3A5 expressers had lower C0 at 48 h (3.7 vs. 6.2 ng/mL, p = 0.03) and at 7 days (8.6 vs. 11.4 ng/mL, p = 0.04) after Tac initiation, tended to take longer to reach Tac therapeutic range (11.8 vs. 8.9 days, p = 0.16), and had higher incidence of both global (92.3% vs. 38.8%, p < 0.001) and grade II-IV acute GVHD (61.5% vs. 24.5%, p = 0.008). These results support the adoption of preemptive pharmacogenetic testing to better predict individual Tac initial dose, helping to achieve the therapeutic range and reducing the risk of acute GVHD earlier.
... Higher tacrolimus concentrations during the first week after allografting with a reduced-intensity conditioning regimen were associated with significantly reduced risk of grade ΙΙ-ΙV aGVHD without increasing risk of relapse. This association was driven by a lower risk of grade ΙΙ-iV aGvHD in patients with week 1 tacrolimus concentrations > 12 ng/ml (24) . ...
Article
Aim: Successful treatment with tacrolimus to prevent graft versus host disease (GVHD) and minimize tacrolimus-related toxicities among allogeneic hematopoietic cell transplantation (alloHCT) recipients is contingent upon quickly achieving and maintaining concentrations within a narrow therapeutic range. The primary objective was to investigate associations between CYP3A4, CYP3A5 or ABCB1 genotype and the proportion of patients that attained an initial tacrolimus goal concentration following initiation of intravenous (iv.) and conversion to oral administration. Materials & methods: We retrospectively evaluated 86 patients who underwent HLA-matched (8/8) related donor alloHCT and were prescribed a tacrolimus-based regimen for GVHD prophylaxis. Results & conclusion: The findings of the present study suggests that CYP3A5 genotype may impact attainment of initial therapeutic tacrolimus concentrations with oral administration in alloHCT recipients.
Article
Full-text available
目的 探讨在异基因造血干细胞移植(allo-HSCT)患者中,CYP3A5基因多态性与他克莫司血药浓度及急性移植物抗宿主病(GVHD)间的关系。 方法 回顾性分析2019年7月至2020年2月在中国医学科学院血液病医院接受allo-HSCT的35例中国成人患者。移植前采集骨髓进行CYP3A5基因分型。应用静脉输注他克莫司、短疗程甲氨蝶呤(MTX)±吗替麦考酚酯进行GVHD预防。在他克莫司用药第2天或第3天监测初始血药浓度,随后每周监测2~3次。根据目标血药浓度(10~15 ng/ml)调整药物剂量。 结果 16例携带CYP3A5*3/*3基因的allo-HSCT患者的初始他克莫司血药浓度(9.82 ng/ml对8.53 ng/ml)、初始血药浓度/剂量(C/D)比值(5.72 ng·ml⁻¹·mg⁻¹对4.26 ng·ml⁻¹·mg⁻¹)、allo-HSCT后第一和第二周C/D比值中位数(5.29 ng·ml⁻¹·mg⁻¹对4.61 ng·ml⁻¹·mg⁻¹,5.65 ng·ml⁻¹·mg⁻¹对4.56 ng·ml⁻¹·mg⁻¹)均明显高于19例至少携带一个CYP3A5*1等位基因的患者(P值分别为0.028、0.001、0.037、0.045)。至少携带一个CYP3A5*1等位基因的患者,allo-HSCT后Ⅲ~Ⅳ级急性GVHD的发生率有高于携带CYP3A5*3/*3基因患者的趋势[(26.3±10.1)%对(6.2±6.1)%,P=0.187]。 结论 CYP3A5基因型导向给药可能有助于allo-HSCT后更快地达到他克莫司目标血药浓度,减少严重急性GVHD的发生,改善移植疗效。
Article
The incidence of graft-versus-host disease (GVHD) after cord blood (CB) transplantation (CBT) is lower than expected given the marked degree of human leukocyte antigen (HLA)-mismatch of CB grafts. While the exact mechanism that underlies this biology remains unclear, it is hypothesized to be due to the low number of mostly immature T-cells infused as part of the graft1,2, and increased tolerance of CB-derived lymphocytes induced by the state of pregnancy. Nevertheless, acute GVHD (aGVHD) is a significant complication of CBT. In contrast, the incidence of chronic GVHD (cGVHD) following CBT is lower than what is observed following matched related or unrelated donor HSC transplantation (HSCT)3-6. This review outlines the guidelines for the prevention and management of acute and chronic GVHD following CBT.
Article
Full-text available
Acute graft-versus-host disease (GVHD) remains a leading cause of morbidity and non-relapse mortality following allogeneic hematopoietic cell transplantation. The clinical staging of GVHD varies greatly between transplant centers and is frequently not agreed upon by independent reviewers. The lack of standardized approaches to handle common sources of discrepancy in GVHD grading likely contributes to why promising GVHD treatments reported from single centers have failed to show benefit in randomized multi-center clinical trials. We developed guidelines through international expert consensus opinion to standardize the diagnosis and clinical staging of GVHD for use in a large international GVHD research consortium. During the first year of use, the guidance was following discussion of complex clinical phenotypes by experienced transplant physicians and data managers. These guidelines increase the uniformity of GVHD symptom capture which may improve the reproducibility of GVHD clinical trials after further prospective validation.
Article
Full-text available
Only a few studies in children have evaluated the efficacy of prophylactic regimens using Tacrolimus on acute graft-versus-host-disease (aGVHD). As a result, optimal Tacrolimus levels in children following matched sibling donor (MSD) allogeneic hematopoietic cell transplantation (AlloHCT) are not well defined. We measured the association between sub therapeutic levels (<10 ng/mL) during weeks 1-4 post-AlloHCT, and the cumulative incidence of grade II-IV aGVHD in children. Additionally, we identified optimal lower cutoff levels for Tacrolimus. Sixty patients received Tacro/MMF between March 2003 and September 2012. The median age was 8 years. Twenty three had a malignant disease and 37 had nonmalignant disorders. The stem cell source included peripheral blood stem cells (n=12) and bone marrow or cord blood (n=48). Conditioning regimen varied. Specifically, 38.3% received a myeloablative regimen, 36.7% receiving a reduced toxicity regimen, and 25% receiving a reduced intensity regimen. Tacrolimus was initiated at 0.03 mg/kg/day via continuous IV infusion, or 0.12mg/kg/d orally. The dose was adjusted to maintain daily steady state concentration within a range 10-20 ng/mL. The overall incidence of grade II-IV aGVHD was 33.3%. On multivariate analysis, a mean Tacrolimus level < 10 ng/mL during week 3 (p=0.042, CI95 1.051-14.28) was significantly associated with increased incidence of grade II-IV aGVHD. Using weeklyreceiver operator curves, the optimal lower cutoff for Tacrolimus levels was 10-11.2 ng/mL. Further prospective studies are warranted to study the incidence of aGVHD comparing the conventional Tacrolimus levels of 5-15 vs. 10-15 ng/ml. Copyright © 2014 American Society for Blood and Marrow Transplantation. Published by Elsevier Inc. All rights reserved.
Article
Full-text available
Because the outcome of allogeneic hematopoietic cell transplantation (HCT) is predominantly influenced by disease type and status, it is essential to be able to stratify patients undergoing HCT by disease risk. The Disease Risk Index (DRI) was developed for this purpose. In this study, we analyzed 13,131 patients reported to the Center for International Blood and Marrow Transplant Research who underwent HCT between 2008 and 2010. The DRI stratified patients into 4 groups with 2-year overall survival (OS) ranging from 64% to 24% and was the strongest prognostic factor, regardless of age, conditioning intensity, graft source, or donor type. A randomly selected training subgroup of 9849 patients was used to refine the DRI, using a multivariable regression model for OS. This refined DRI had improved prediction ability for the remaining 3282 patients compared with the original DRI or other existing schemes. This validated and refined DRI can be used as a 4- or 3-group index, depending on the size of the cohort under study, for prognostication; to facilitate the interpretation of single-center, multicenter, or registry studies; to adjust center outcome data; and to stratify patients entering clinical trials that enroll patients across disease categories.
Article
Full-text available
The calcineurin inhibitor tacrolimus is the backbone of immunosuppressive drug therapy after solid organ transplantation. Tacrolimus is effective in preventing acute rejection but has considerable toxicity and displays marked inter-individual variability in its pharmacokinetics and pharmacodynamics. The genetic basis of these phenomena is reviewed here. With regard to its pharmacokinetic variability, a single nucleotide polymorphism (SNP) in cytochrome P450 (CYP) 3A5 (6986A>G) has been consistently associated with tacrolimus dose requirement. Patients expressing CYP3A5 (those carrying the A nucleotide, defined as the *1 allele) have a dose requirement that is around 50 % higher than non-expressers (those homozygous for the G nucleotide, defined as the *3 allele). A randomised controlled study in kidney transplant recipients has demonstrated that a CYP3A5 genotype-based approach to tacrolimus dosing leads to more patients reaching the target concentration early after transplantation. However, no improvement of clinical outcomes (rejection incidence, toxicity) was observed, which may have been the result of the design of this particular study. In addition to CYP3A5 genotype, other genetic variants may also contribute to the variability in tacrolimus pharmacokinetics. Among these, the CYP3A4*22 and POR*28 SNPs are the most promising. Individuals carrying the CYP3A4*22 T-variant allele have a lower tacrolimus dose requirement than individuals with the CYP3A4*22 CC genotype and this effect appears to be independent of CYP3A5 genotype status. Individuals carrying the POR*28 T-variant allele have a higher tacrolimus dose requirement than POR*28 CC homozygotes but this association was only found in CYP3A5-expressing individuals. Other, less well-defined SNPs have been inconsistently associated with tacrolimus dose requirement. It is envisaged that in the future, algorithms incorporating clinical, demographic and genetic variables will be developed that will aid clinicians with the determination of the tacrolimus starting dose for an individual transplant recipient. Such an approach may limit early tacrolimus under-exposure and toxicity. With regard to tacrolimus pharmacodynamics, no strong genotype-phenotype relationships have been identified. Certain SNPs associate with rejection risk but these observations await replication. Likewise, the genetic basis of tacrolimus-induced toxicity remains unclarified. SNPs in the genes encoding for the drug transporter ABCB1 and the CYP3A enzymes may relate to chronic nephrotoxicity but findings have been inconsistent. No genetic markers reliably predict new-onset diabetes mellitus after transplantation, hypertension or neurotoxicity. The CYP3A5*1 SNP is currently the most promising biomarker for tailoring tacrolimus treatment. However, before CYP3A5 genotyping is incorporated into the routine clinical care of transplant recipients, prospective clinical trials are needed to determine whether such a strategy improves patient outcomes. The role of pharmacogenetics in tacrolimus pharmacodynamics should be explored further by the study of intra-lymphocyte and tissue tacrolimus concentrations.
Article
Full-text available
We studied whether early CsA trough levels were associated with the risk of acute GVHD in 337 patients after either sibling PBSC or double umbilical cord blood transplantation. All patients, regardless of donor type, started CsA at a dose of 5 mg/kg i.v. divided twice daily, targeting trough concentrations 200-400 ng/mL. The CsA level was studied by a weighted average method calculated by giving 70% of the weight to the level that was measured just before the onset of the event or day +30. We found that higher weighted average CsA trough levels early post transplantation contributed to lower risk of acute GVHD, and lower non-relapse and overall mortality. Thus, our data support close monitoring with active adjustments of CsA dosing to maintain therapeutic CsA levels in the first weeks of allo-HCT. In patients who are near or even modestly above the CsA target trough level, in the absence of CsA-related toxicity, dose reduction should be cautious to avoid subtherapeutic drug levels resulting in higher risk of acute GVHD.Bone Marrow Transplantation advance online publication, 16 September 2013; doi:10.1038/bmt.2013.139.
Article
AML is currently the most common indication for reduced-intensity conditioned (RIC) allo-SCT. Reduced-intensity regimens allow a potent GVL response to occur with minimized treatment-related toxicity in patients of older age or with comorbidities that preclude the use of myeloablative conditioning. Whether RIC SCT is appropriate for younger and more standard risk patients is not well defined and the field is changing rapidly; a prospective randomized trial of myeloablative vs RIC (BMT-CTN 0901) was recently closed when early results indicated better outcomes for myeloablative regimens. However, detailed results are not available, and all patients in that study were eligible for myeloablative conditioning. RIC transplants will likely remain the standard of care as many patients with AML are not eligible for myeloablative conditioning. Recent publication of mature results from retrospective and prospective cohorts provide contemporary efficacy and toxicity data for these attenuated regimens. In addition, recent studies explore the use of alternative donors, introduce regimens that attempt to reduce toxicity without reducing intensity, and identify predictive factors that pave the way to personalized approaches. These studies paint a picture of the future of RIC transplants. Here we review the current status of RIC allogeneic SCT in AML.Bone Marrow Transplantation advance online publication, 2 March 2015; doi:10.1038/bmt.2015.7.
Article
There have been no randomized trials that have compared peripheral blood (PB) with bone marrow (BM) grafts in the setting of reduced-intensity conditioning (RIC) transplantations for hematologic malignancy. Because immune modulation plays a significant role in sustaining clinical remission after RIC, we hypothesize that higher graft-versus-host disease (GVHD) associated with PB transplantation may offer a survival advantage. The primary outcome evaluated was overall survival. Cox regression models were built to study outcomes after transplantation of PB (n = 887) relative to BM (n = 219) for patients with acute myeloid leukemia, myelodysplastic syndrome, or non-Hodgkin lymphoma, the three most common indications for unrelated RIC transplantation. Transplantations were performed in the United States between 2000 and 2008. Conditioning regimens consisted of an alkylating agent and fludarabine, and GVHD prophylaxis involved a calcineurin inhibitor (CNI) with either methotrexate (MTX) or mycophenolate mofetil (MMF). After adjusting for age, performance score, donor-recipient HLA-match, disease, and disease status at transplantation (factors associated with overall survival), there were no significant differences in 5-year rates of survival after transplantation of PB compared with BM: 34% versus 38% with CNI-MTX and 27% versus 20% with CNI-MMF GVHD prophylaxis. Survival after transplantation of PB and BM are comparable in the setting of nonirradiation RIC regimens for hematologic malignancy. The effect of GVHD prophylaxis on survival merits further evaluation. © 2014 by American Society of Clinical Oncology.
Article
Graft-versus-host disease (GVHD) represents the most serious and challenging complication of allogeneic haematopoietic stem-cell transplantation (HSCT). New insights on the role of regulatory T-cells, T cells, and antigen-presenting cells have led to an improved understanding of the pathophysiology of GVHD. However, little progress has been made since the introduction of calcineurin-inhibitor-based regimens in the mid-1980s. Despite standard prophylaxis with these regimens, GVHD still develops in approximately 40-60% of recipients. Thus, there is a need for developing newer approaches to mitigate GVHD, which may facilitate the use of allogeneic HSCT for the treatment of a wider range of haematological cancers. We discuss the rationale, clinical evidence, and outcomes of current (and widely employed) strategies for GVHD prophylaxis, namely calcineurin-inhibitor-based regimens (such as cyclosporine or tacrolimus) combined with methotrexate or mycophenolate mofetil. We assess the clinical evidence for emerging approaches in the prevention of GVHD, including therapies targeting T cells or B cells, the use of mesenchymal stem cells, chemo-cytokine antagonists (such as maraviroc, TNF-α inhibitor, IL-2 receptor antagonist, IL-6 inhibitor), and the use of novel molecular regulators that target multiple cell types simultaneously, including atorvastatin, bortezomib, and epigenetic modulators.
Article
Allogeneic hematopoietic stem cell transplantation (alloSCT) is a potentially curative therapy for many hematologic and immunologic diseases. Further, partial or full donor hematopoietic chimerism following alloSCT may be sufficient to guarantee immunologic tolerance to solid organs from the same donor, obviating any requirement for prolonged pharmacologic immunosuppression. Despite alloSCT's potential, the procedure is beset by two major limitations. The first relates to the procedure's toxicity, including conditioning regimen toxicity, graft-versus-host disease (GVHD), and infection. The second limitation is the lack of histocompatible donors. A human leukocyte antigen (HLA)-matched sibling or unrelated donor cannot be identified expeditiously for up to 40% of patients. Historically, alloSCT from partially HLA-mismatched, or HLA-haploidentical, relatives has been complicated by unacceptably high incidences of graft rejection, severe GVHD, and non-relapse mortality. Recently, our groups have developed a method to selectively deplete alloreactive cells in vivo by administering high doses of cyclophosphamide in a narrow window after transplantation. Using high-dose, post-transplantation cyclophosphamide (PT/Cy), crossing the HLA barrier in alloSCT is now feasible and donors can be found for nearly all patients. This review discusses the history of HLA-haploidentical SCT, recent clinical results, and immunologic mechanisms of action of high-dose PT/Cy for prevention of graft rejection and GVHD.