You are on page 1of 15

ORIGINAL PAPER

e-ISSN 2329-0358
© Ann Transplant, 2024; 29: e942167
DOI: 10.12659/AOT.942167

Received:
Accepted:
2023.08.14
2024.02.09 Long-Term Outcomes with Prolonged-
Available online:
Published:
2024.03.07
2024.03.19 Release Tacrolimus in Kidney Transplantation:
A Retrospective Real-World Data Analysis

Authors’ Contribution: ABDE 1 Wilfried Gwinner 1 Department of Nephrology, Hannover Medical School, Hannover, Germany
Study Design A ACDE 2 Swapneel Anaokar 2 Astellas Pharma Europe Ltd., Addlestone, United Kingdom
Data Collection B 3 Astellas Pharma GmbH, Munich, Germany
Statistical Analysis C ACDE 2 Martin Blogg 4 Department of Nephrology and Hypertension, Universitätsklinikum Erlangen
Data Interpretation D ACDE 3 Birgit Hermann und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
Manuscript Preparation E CDE 2 Carola del Pilar Repetur
Literature Search F
Funds Collection G ABDE 4 Mario Schiffer

Corresponding Author: Wilfried Gwinner, e-mail: gwinner.wilfried@mh-hannover.de


Financial support: This study was funded by Astellas Pharma, Inc. Medical writing and editorial support were funded by Astellas Pharma, Inc.
Conflict of interest: All authors report non-financial support from Astellas during the conduct of the study. WG received consulting fees from Novartis
and Bayer. MS received an unrestricted grant from Novartis, and consulting fees for advisory boards from Novartis and Bayer. SA,
MB, and CPR are employees of Astellas Pharma Europe. BH is an employee of Astellas Pharma GmbH

Background: Long-term real-world outcomes data for kidney transplant recipients (KTRs) converting from immediate-release
tacrolimus (IRT) to prolonged-release tacrolimus (PRT) are limited.
Material/Methods: A retrospective, non-interventional review of adult KTRs treated with PRT for ³1 month was conducted in
Germany. Data were extracted from time of transplant (2008-2014) to 2018. Primary composite endpoints
(graft loss, biopsy-confirmed acute rejection, graft dysfunction) and secondary endpoints (all-cause mortality,
kidney function course, and tacrolimus dose/trough levels) were analyzed for sub-cohorts: de novo PRT, early
conversion from IRT (within 6 months post-transplant), and late conversion (7 months to 3 years).
Results: Analysis included 163 patients (101 de novo, 12 early converters, and 50 late converters). The overall Kaplan-Meier
estimate of freedom from efficacy failure through 5 years was 0.537, (95% confidence interval (CI) 0.455-0.612)
(de novo: 0.512 [0.407-0.608]; early converters: 0.500 [0.208-0.736]; late converters: 0.594 [0.443-0.717]). The
overall survival rate was 0.925 (95% CI 0.872-0.957) (de novo: 0.900 [0.823-0.945]; early converters: 0.917
[0.539-0.988]; late converters: 0.977 [0.846-0.997]). During follow-up, there was a gradual reduction in tacro-
limus dose and trough levels; kidney function remained stable in all cohorts. Multivariable analysis found re-
transplantation, organ donor quality, best estimated glomerular filtration rate 8-12 weeks after transplant, and
treatment center (between-center differences in age, sex, donor status/quality) were significantly associated
with efficacy failure.
Conclusions: There was no difference in long-term survival profiles between KTRs who received PRT de novo vs those who
converted from IRT, with 5-year survival remaining high in both groups.
Keywords: Immunosuppression Therapy • Tacrolimus • Kidney Transplantation • Retrospective Studies •
Clinical Study
Abbreviations: BCAR – biopsy-confirmed acute rejection; CKD-EPI – Chronic Kidney Disease Epidemiology Collaboration;
CMV – cytomegalovirus; CI – confidence interval; ECD – expanded criteria donor; eGFR – estimated glo-
merular filtration rate; FAC – full analysis cohort; HLA – human leukocyte antigen; HR – hazard ratio;
IgG – immunoglobulin; IRT – immediate-release tacrolimus; KM – Kaplan-Meier; KTR – kidney transplant
recipient; MDRD-4 – modification of diet in renal disease-4; NE – not estimable; PRT – prolonged-release
tacrolimus; SCD – standard criteria donor; SD – standard deviation; SE – standard error

Full-text PDF: https://www.annalsoftransplantation.com/abstract/index/idArt/942167

4124   9   5   34

Publisher’s note: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated
organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be
made by its manufacturer, is not guaranteed or endorsed by the publisher

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-1 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
ORIGINAL PAPER Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167

Introduction graft loss and a 5-year Kaplan-Meier (KM) estimated rejection


rate of 26.9% [23]. In the ADMIRAD follow-up study, a retro-
Despite advances in immunosuppressive therapy and im- spective non-interventional chart review of KTRs treated with
provements in 1-year transplant survival rates over several PRT following conversion from IRT, the KM survival rate with-
decades [1,2], long-term survival after kidney transplantation out efficacy failure (defined as transplantation, nephrectomy,
remains challenging, with data from the US and Europe sug- death or return to dialysis) from randomization to year 5 was
gesting that 10-year graft survival is less than 60% [3]. 74.1%, and overall patient survival was 98.1% and 88.0% at
5 and 10 years after transplant, respectively [25].
Tacrolimus is the mainstay of post-kidney transplant immuno-
suppression, commonly used with other immunosuppressants There is a need to further characterize long-term clinical out-
as dual- or triple-drug regimens to prevent acute kidney rejec- comes with PRT in both de novo patients and those converting
tion [4,5]. While short-term patient and graft survival rates af- from IRT. The aim of this retrospective study was to assess the
ter kidney transplant are high, longer-term graft survival may be long-term outcomes of PRT-based immunosuppressive ther-
jeopardized by nonadherence to immunosuppressive regimens apy in de novo KTRs or those converting from IRT to PRT in a
[5,6]. A prospective study following kidney transplant recipients real-world clinical practice setting.
(KTRs) for 18 years found nonadherence to be a major cause of
graft loss, with most nonadherence occurring 3 years or more af-
ter transplantation [6]. For tacrolimus, which has a narrow thera- Material and Methods
peutic index, adherence and maintenance of stable blood levels
are important [5]. Variability in systemic tacrolimus exposure is Study Design and Population
a risk factor for graft loss in the early and late post-transplanta-
tion period [7,8] and reducing intra-patient variability in tacroli- This was a retrospective, non-interventional, observational chart
mus exposure has been shown to improve graft survival [9,10]. review conducted at 2 kidney transplant centers in Germany:
Universitätsklinikum Erlangen (Center 1) and Medizinische
In addition to the traditional twice-daily immediate-release ta- Hochschule Hannover (Center 2).
crolimus (IRT) formulation, a once-daily prolonged-release ta-
crolimus (PRT) formulation has been developed to provide a Patients were included if they were ³18 years of age, had re-
more consistent level of total tacrolimus exposure and more ceived a kidney transplant between 2008 and 2014 from a de-
convenient dosing regimen [5,11-13]. Benefits of PRT over IRT ceased or living donor, and had received PRT-based therapy
include improved patient adherence [14] and reduced intra-pa- (Advagraf®, Astellas Pharma Europe BV, Leiden, the Netherlands)
tient variability in tacrolimus exposure [11,12], both of which for ³1 month either de novo or after conversion from IRT
are determinants of long-term graft outcome, with no differ- (Prograf®, Astellas Pharma Europe BV, Leiden, the Netherlands).
ence in exposure [7,8,15-19]; however, there could be some for- PRT-based therapy was defined as PRT at a strength of 0.5 mg,
mulation and pharmacokinetic differences between PRT med- 1 mg, 3 mg, and 5 mg; capsules were administered orally once
ications. Additionally, the benefit of switching to PRT may be daily, either de novo or after conversion from IRT. Patients were
impacted by the exact formulation used; recent data showed excluded if they had contraindications for tacrolimus treat-
differences in intestinal absorption between 2 formulations ment, had received IRT >3 years prior to conversion to PRT,
of PRT, leading to differences in variability of exposure [20]. had switched from a non-tacrolimus-based immunosuppres-
sive regimen, or had received a multi-organ transplant. Patients
The efficacy and safety of PRT is well established with respect to could have received a kidney from a deceased or living donor.
short-term outcomes, in both de novo KTRs and those switch- Minimum thresholds for inclusion based on estimated glomer-
ing from IRT [21]. Longer-term outcomes with PRT are less well ular filtration rate (eGFR) at baseline were not defined.
characterized, but the limited trial data that are available are
encouraging [22,23]. A 5-year, non-interventional, prospec- The study was conducted in compliance with national and
tive follow-up study of KTRs randomized to de novo PRT in European Union requirements for ensuring the rights of par-
the Phase IV, open-label, ADHERE (1-year) study revealed pa- ticipants in non-interventional studies and followed the reg-
tient and graft survival rates of 91.9% and 89.3%, respectively ulations of the Declaration of Helsinki and the Declaration of
[22]. Similar 5-year patient and graft survival rates (94.4% and Istanbul. Approval was obtained from the Institutional Ethics
88.1%) were seen in a follow-up of KTRs receiving de novo PRT Commission at both centers. Patients who were alive at the
in the Phase IV, open-label, 24-week ADVANCE trial [24]. In a time of data extraction provided written informed consent.
5-year open-label, single-center, prospective study of KTRs in The requirement for informed consent was waived for patients
Japan, the patient survival rate with de novo PRT was 96.7% who had died before the start of the study. The reporting of
at 5 years after transplantation, with 6.8% of patients having this study conforms to the STROBE statement [26].

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-2 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167
ORIGINAL PAPER

Data Extraction • Late conversion – received de novo IRT and subsequently con-
verted to PRT between 7 months and 3 years post-transplant.
Data were sourced from the medical charts of transplant re-
cipients at Center 1 and Center 2. For each eligible patient, Post hoc analyses included comparison of the primary endpoint
retrospective data from time of transplant (2008-2014) until (and individual components of the composite) between Center
the date of the last follow-up visit prior to December 31, 2018 1 and Center 2 and the association of further variables with
(maximum 11-year period) were extracted from existing medi- the primary endpoint including; cohort (de novo vs combined
cal records by center personnel using an electronic case report conversion [early plus late conversion sub-groups]), baseline
form. Data were recorded as part of routine care and no spe- kidney function (8-12 weeks after transplantation), and the im-
cific patient visits were required as part of the study. Collected pact on kidney function in patients who had converted from
data included: date of PRT initiation or conversion from IRT; IRT (combined conversion sub-group) versus the de novo PRT
reason for conversion to PRT from IRT; drug dose; tacrolimus sub-group. Post-transplant prescribed tacrolimus doses and
blood trough level; date of graft loss and type of graft loss (re- trough levels from time of transplant were also assessed in
transplantation, death, or first day of dialysis); date of biopsy- the FAC and combined conversion cohorts.
confirmed acute rejection (BCAR); date of occurrence of graft
dysfunction, defined as eGFR of <40 mL/min/1.73 m2 accord- Statistical Analysis
ing to the modification of diet in renal disease-4 (MDRD-4) for-
mula; renal parameters from laboratory assessment, includ- Continuous data were reported as mean (standard devia-
ing serum creatinine, Chronic Kidney Disease Epidemiology tion [SD]), median (lower quartile [Q1], upper quartile [Q3]).
Collaboration (CKD-EPI) and baseline eGFR, defined as best Categorical data were reported as frequencies and percent-
eGFR 8-12 weeks post-transplant (MDRD-4); and cause of pa- ages. Patients with missing data were not excluded, and each
tient death. Missing data were requested from the treating variable was summarized based on the available data. Primary
nephrologists by center personnel. and secondary endpoint and post hoc analyses were conduct-
ed on all study cohorts.
Endpoints
Analysis of the primary composite endpoint provided KM es-
The primary composite endpoint of efficacy failure was the ear- timates and 95% confidence intervals (95% CIs) of the time
liest occurrence of any of the following: (1) graft loss, includ- from transplant to the first incidence of the components of the
ing re-transplantation, death or dialysis; (2) BCAR, determined endpoint, using Greenwood’s formula to estimate the standard
by the investigator and regarded as acute antibody-mediated error of survival. Statistical significance was determined using
rejection (grade I, II or III) or acute T-cell-mediated rejection log-rank testing of all events captured over the follow-up pe-
(grade IA, IB, IIA, IIB or III), according to Banff criteria valid at riod. KM estimates (with 95% CI, calculated as defined above)
the time of biopsy [27]; or (3) graft dysfunction, defined previ- of the composite endpoint at 5 years (the primary objective)
ously. Patients who never had eGFR ³40 mL/min/1.73 m2 were and for each year of follow-up were calculated, as well as the
considered to have graft dysfunction on the day of first occur- estimated median time to composite endpoint (with 95% CI).
rence of eGFR <40 mL/min/1.73 m2, which could be on day 1. Patients who were lost to follow-up or were alive at the end of
the study without occurrence of the endpoint were censored
Secondary endpoints included all-cause mortality, kidney func- on the day of the last available follow-up visit. For the sec-
tion over time, and prescribed tacrolimus dosing and trough ondary endpoints, all-cause mortality was analyzed using KM
levels. In addition, exploratory analyses assessed independent methods as per the primary composite endpoint. Other second-
risk factors for impact on the composite endpoint. ary endpoints were analyzed using descriptive statistics, with
baseline measurements performed on the date of transplan-
Sub-group and Post Hoc Analyses tation. Post hoc analyses compared the KM estimates of free-
dom from efficacy failure between the de novo and combined
Primary analyses of endpoints were conducted for the full anal- conversion (early plus late) cohorts and the primary composite
ysis cohort (FAC), comprising all eligible patients who received endpoint by eGFR at baseline (8-12 weeks post-transplant) us-
³1 month of continuous PRT following kidney transplantation, ing fixed thresholds for eGFR categories (<30 mL/min/1.73 m2,
and 3 sub-groups as follows: ³30-<45 mL/min/1.73 m2, ³45-<60 mL/min/1.73 m2, and ³60
• De novo – PRT treatment initiated within the first month mL/min/1.73 m2). Descriptive graphs plotting means and stan-
post-transplant dard errors of eGFR over time from time of transplant were plot-
• Early conversion – received de novo IRT and subsequently ted for the de novo and combined conversion cohort. Patients
converted to PRT within 6 months post-transplant with graft loss were included in the analysis of subsequent vis-
its and assigned an eGFR of 0. Sensitivity analyses, conducted

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-3 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
ORIGINAL PAPER Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167

to test the robustness of the primary composite endpoint, in-


cluded KM estimates of the individual components of the com- Eligible in site databases, n=243
posite endpoint and patients with and without BCAR or kid-
ney dysfunction prior to conversion. Not participated, n=80
• Not reached, n=17
• Not consented, n=62
In the exploratory analysis, independent risk factors were • Died before consenting, n=1
modeled using a Cox regression model adjusting for potential
confounders. Univariate models were used to determine the Included in analyses, n=163
• Consented, n=137
variables associated with PRT effectiveness and a final mul- • Included without consent (died), n=26
tivariable model was developed to quantify the contribution
of each variable to the between-patient variation in outcome.
The models used eGFR according to the MDRD-4 formula. The Figure 1. Patient flow through the study.
Cox multivariable models (exploratory and post hoc analyses)
used forward selection (P value threshold 0.05) due to con- median time to primary endpoint event was 7.11 years (95% CI
vergence issues observed for the models that included all po- 3.87-not estimable [NE]). There was no clear difference in the
tential covariates. All variables with P<0.2 in univariate Cox re- 5-year time-to-event profile between the cohorts (log-rank test
gression were considered for model entry. Statistical analyses P=0.744) and the KM estimate of freedom from efficacy fail-
were performed using SAS® software, version 9.4.1 or higher. ure through 5 years was 0.537 (95% CI 0.455-0.612) (Table 4).

Sensitivity Analyses of the Primary Composite Endpoint


Results
The KM estimates of individual components of the primary
Among 243 patients eligible for the study, 163 met the inclu- composite endpoint are presented only as a sensitivity analy-
sion criteria and were included in the FAC (Figure 1). More pa- sis owing to potential bias due to the competing risks of occur-
tients were treated with PRT de novo (n=101) than converted rence of the other primary endpoint components. When each
from IRT (n=62; early n=12, late n=50). The majority (71.8%) component was analyzed individually, there was no major dif-
of patients were male and the median (Q1-Q3) age was 52.0 ference between the de novo, early conversion, and late con-
(42-62) years (Table 1). In the de novo and early conversion version cohorts (Table 5). The survival rate without experienc-
cohorts, around three-quarters of transplants were from de- ing graft loss in the FAC was 0.862 (95% CI 0.794-0.909) at 5
ceased donors, whereas the late conversion cohort had a com- years. When combining early and late conversion cohorts into
paratively higher proportion of living donors (56% living/44% a single ‘conversion’ cohort in the post hoc analysis, there was
deceased). Furthermore, patients in the late conversion cohort also no significant difference between the cohorts (log-rank test
had a median (Q1-Q3) time from transplant to conversion of P=0.603; Figure 3). The KM estimate of freedom from efficacy
21.41 (15-27) months. Median (Q1-Q3) time from initiation failure through 5 years was 0.512 (95% CI 0.407-0.608) in the
of PRT to discontinuation, censoring or loss-to-follow-up was de novo PRT cohort and 0.576 (95% CI 0.442-0.689) in the com-
55.26 (33.58-85.19) months. Significant between-center dif- bined (early plus late) conversion cohort. The crude incidence of
ferences in baseline patient and donor characteristics were primary endpoint event by the end of the study (date of the last
evident. Patients at Center 1 were older and included a high- available follow-up visit) was 48.5% (49/101) in the de novo PRT
er proportion with comorbidities, a lower proportion of trans- cohort and 43.5% (27/62) in the combined conversion cohort.
plants from living donors, and a higher proportion with ex-
panded criteria donor (ECD) status (Table 2). There were 22 patients in the primary analysis who had graft
dysfunction or BCAR prior to conversion to PRT (17 of whom
Primary Composite Endpoint were included in the late conversion cohort). Sensitivity anal-
ysis of the primary composite endpoint for the remaining 141
Overall, 76 (46.6%) patients had a primary composite end- patients after exclusion of these patients revealed a KM esti-
point event (de novo 49 [48.5%]; early converters 6 [50.0%]; mate of freedom from efficacy failure through 5 years of 0.622
late converters 21 [42.0%]), and 87 (53.4%) patients were (95% CI 0.533-0.698).
censored. Post hoc analysis revealed 60.5% of patients had
a primary composite endpoint event at Center 1 vs 32.9% at Survival
Center 2 (Table 3).
Overall, 18 (11.0%) patients died over the maximum post-
KM estimates of time from transplant to first primary composite transplant study period (11 years). The KM estimate of surviv-
endpoint event are presented in Figure 2. The KM estimate of al rate at 5 years was 0.925 (95% CI 0.872-0.957) in the FAC

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-4 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167
ORIGINAL PAPER

Table 1. Baseline characteristics of patients.

Cohort
Parameter De novo Early conversion Late conversion Full analysis
(n=101) (n=12) (n=50) (N=163)

Study site, n (%)

Center 1 57 (56.4) 7 (58.3) 17 (34.0) 81 (49.7)

Center 2 44 (43.6) 5 (41.7) 33 (66.0) 82 (50.3)

Age, years

Mean (SD) 51.3 (13.7) 53.1 (13.9) 48.3 (13.3) 50.5 (13.6)

53.0 49.0 51.0 52.0


Median (Q1-Q3)
(42.0-63.0) (42.5-63.5) (40.0-58.0) (42.0-62.0)

Age group, n (%)

³18 to <65 years 81 (80.2) 9 (75.0) 45 (90.0) 135 (82.8)

³65 years 20 (19.8) 3 (25.0) 5 (10.0) 28 (17.2)

Sex, n (%)

Male 71 (70.3) 9 (75.0) 37 (74.0) 117 (71.8)

Female 30 (29.7) 3 (25.0) 13 (26.0) 46 (28.2)

Race/ethnicity, n (%)

White/Caucasian 55 (54.5) 8 (66.7) 43 (86.0) 106 (65.0)

Black/African-American/Caribbean 1 (1.0) 1 (8.3) 1 (2.0) 3 (1.8)

Middle Eastern 1 (1.0) 1 (2.0) 2 (1.2)

Other 2 (2.0) 2 (1.2)

Not available 42 (41.6) 3 (25.0) 5 (10.0) 50 (30.7)

Number of previous transplants, n (%)

0 100 (99.0) 8 (66.7) 45 (90.0) 153 (93.9)

1 1 (1.0) 4 (33.3) 4 (8.0) 9 (5.5)

2 1 (2.0) 1 (0.6)

Donor vital status

Living 24 (23.8) 3 (25.0) 28 (56.0) 55 (33.7)

Deceased 77 (76.2) 9 (75.0) 22 (44.0) 108 (66.3)

Time from transplant to PRT conversion, months

Mean (SD) 0.02 (0.09) 3.80 (1.56) 21.02 (7.47) 6.74 (10.43)

0.00 3.67 21.41 0.00


Median (Q1-Q3)
(0.00-0.00) (2.40-5.05) (14.72-26.71) (0.00-12.98)

Time from PRT conversion to discontinuation,


censoring or loss-to-follow-up, months

Mean (SD) 65.22 (35.84) 55.86 (26.12) 42.85 (16.27) 57.67 (31.99)

77.47 52.24 41.27 55.26


Median (Q1-Q3)
(35.94-89.63) (44.81-64.13) (31.97-50.89) (33.58-85.19)

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-5 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
ORIGINAL PAPER Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167

Table 1 continued. Baseline characteristics of patients.

Cohort
Parameter De novo Early conversion Late conversion Full analysis
(n=101) (n=12) (n=50) (N=163)

Reason for conversion to PRT

Adherence issues 2 (4.0) 2 (1.2)

Trough variability 1 (1.0) 4 (33.3) 18 (36.0) 23 (14.1)

Patient preference 1 (1.0) 3 (6.0) 4 (2.5)

Other clinical preference 7 (14.0) 7 (4.3)

Study participation 93 (92.1) 93 (57.1)

Not available 6 (5.9) 8 (66.7) 20 (40.0) 34 (20.9)

PRT – prolonged-release tacrolimus; Q – quartile; SD – standard deviation. Center 1, Universitätsklinikum Erlangen;


Center 2, Medizinische Hochschule Hannover.

Table 2. Patient demographics per transplant center.

Center 1 Center 2
Parameter P value
(n=81) (n=82)

Age, mean (SD) 54.6 (12.3) 46.6 (13.7) <0.001

Age ³65 years, n (%) 20 (24.7) 8 (9.8) 0.011

Male, n (%) 62 (76.5) 55 (67.1) 0.179

Living donor, n (%) 20 (24.7) 35 (42.7) 0.015

ECD status, n (%) 36 (44.4) 9 (11.0) <0.001

Comorbidities at baseline (type 2 diabetes), n (%) 18 (22.2) 6 (7.3) 0.007

CMV donor IgG positive/recipient IgG negative, n (%) 25 (30.9) 19 (23.2) 0.269

CMV – cytomegalovirus; ECD – expanded criteria donor; IgG – immunoglobulin G; SD – standard deviation. Center 1, Universitäts-
klinikum Erlangen; Center 2, Medizinische Hochschule Hannover.

Table 3. Post hoc analysis of outcomes per transplant center.

Center 1 Center 2
Parameter
(n=81) (n=82)

Endpoint, n (%)

Primary composite endpoint 49 (60.5) 27 (32.9)

Graft loss 20 (24.7) 6 (7.3)

BCAR 26 (32.1) 19 (23.2)

Graft dysfunction 35 (43.2) 6 (7.3)

Death 17 (21.0) 1 (1.2)

BCAR – biopsy-confirmed acute rejection. Center 1, Universitätsklinikum Erlangen; Center 2, Medizinische Hochschule Hannover.

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-6 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167
ORIGINAL PAPER

1.0
Proportion of patients surviving without reaching the composite endpoint + Censored

0.8

0.6

0.4

0.2
De novo cohort
Early conversion
Late conversion
All patients

0.0 P value=0.744

0 1 2 3 4 5 6 7 8 9 10 11
Follow-up (years)

De novo cohort, n: 101 67 60 56 48 43 38 31 12 8 6 0


Early conversion, n: 12 7 6 6 6 3 2 1 0
Late conversion, n: 50 35 32 30 27 16 11 3 1 1 0
All patients, n: 163 109 98 92 81 62 51 35 13 9 6 0

Figure 2. Kaplan-Meier plot of time from transplant to primary composite endpoint event in the full analysis cohort and
the 3 tacrolimus conversion cohorts. P-value is based on time to event though 5 years.

and remained high in all cohorts: (0.900 (95% CI 0.823-0.945) Tacrolimus Dose and Trough Levels
for the de novo cohort, 0.917 (95% CI 0.539-0.988) for the ear-
ly conversion cohort, and 0.977 (95% CI 0.846-0.997) for the Tacrolimus doses prescribed over time were captured in a
late conversion cohort (Table 6). descriptive plot stratified by conversion cohorts (Figure 4A),
which shows that mean prescribed doses were similar for the
Impact of Independent Variables on Composite de novo and late conversion cohorts throughout the follow-
Primary Endpoint up period. Mean doses in the early conversion cohort, which
had considerably fewer patients (n=12 vs n=101 and n=50, in
All variables with P<0.2 in the univariate analysis (results shown the de novo and late conversion cohorts, respectively) were
in Table 7) were considered for multivariable model entry. The comparatively lower. The tacrolimus dose curves in all 3 co-
multivariable Cox proportional hazards analysis revealed vari- horts followed a downward gradient, indicating a gradual ta-
ables significantly associated with the primary composite end- pering of dosage over time before the dose became stable.
point (P£0.001) including: number of previous transplants (1 Similarly, in all 3 cohorts, tacrolimus trough levels gradually
vs 0; hazard ratio [HR] 6.592; 95% CI 2.331-18.639); donor decreased over time before whole blood tacrolimus trough lev-
quality (ECD vs standard criteria donor [SCD] status: HR 2.931; els reached a stable level (Figure 4B). At baseline, the mean
95% CI 1.554-5.526); baseline eGFR (best eGFR 8-12 weeks tacrolimus trough levels ranged from 10-12 ng/mL in all co-
post-transplant, time-dependent variable: HR 0.933; 95% CI horts. The mean levels in all cohorts remained generally sim-
0.894-0.974); as well as study site (Center 2 vs Center 1: HR ilar throughout the duration of follow-up.
0.196; 95% CI 0.078-0.494) (Table 7).

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-7 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
ORIGINAL PAPER Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167

Table 4. Kaplan-Meier estimates of freedom from the primary composite endpoint stratified by population cohort (first 5 years
post-transplant).

Cohort
KM rate estimate
Year De novo Early conversion Late conversion Full analysis
(95% CI)
(n=101) (n=12) (n=50) (N=163)

Composite 1 0.691 (0.591-0.772) 0.500 (0.208-0.736) 0.680 (0.532-0.790) 0.673 (0.595-0.739)

2 0.638 (0.535-0.724) 0.500 (0.208-0.736) 0.640 (0.491-0.756) 0.628 (0.549-0.698)

3 0.616 (0.512-0.704) 0.500 (0.208-0.736) 0.620 (0.471-0.738) 0.608 (0.528-0.679)

4 0.547 (0.442-0.641) 0.500 (0.208-0.736) 0.620 (0.471-0.738) 0.567 (0.486-0.640)

5 0.512 (0.407-0.608) 0.500 (0.208-0.736) 0.594 (0.443-0.717) 0.537 (0.455-0.612)

CI – confidence interval; KM – Kaplan-Meier. The KM estimates of individual components are presented only as a sensitivity analysis of
the primary endpoint owing to potential bias due to the competing risks of occurrence of the other primary endpoint components.

Table 5. Sensitivity analysis of primary endpoint: Kaplan-Meier estimates of the individual components of the composite endpoint
stratified by population cohort (first 5 years post-transplant).

Cohort
KM rate estimate
Year
(95% CI) De novo Early conversion Late conversion Full analysis
(n=101) (n=12) (n=50) (N=163)

Graft loss 1 0.959 (0.895-0.985) 1.000 (1.000-1.000) 1.000 (1.000-1.000) 0.975 (0.935-0.991)

2 0.927 (0.852-0.964) 0.917 (0.539-0.988) 0.980 (0.866-0.997) 0.943 (0.892-0.970)

3 0.916 (0.838-0.957) 0.917 (0.539-0.988) 0.960 (0.848-0.990) 0.929 (0.876-0.960)

4 0.868 (0.778-0.923) 0.917 (0.539-0.988) 0.939 (0.822-0.980) 0.894 (0.832-0.934)

5 0.842 (0.748-0.904) 0.815 (0.435-0.951) 0.912 (0.780-0.966) 0.862 (0.794-0.909)

BCAR 1 0.818 (0.726-0.881) 0.583 (0.270-0.801) 0.740 (0.595-0.840) 0.775 (0.702-0.832)

2 0.759 (0.660-0.833) 0.583 (0.270-0.801) 0.720 (0.574-0.824) 0.734 (0.658-0.797)

3 0.759 (0.660-0.833) 0.583 (0.270-0.801) 0.720 (0.574-0.824) 0.734 (0.658-0.797)

4 0.733 (0.630-0.812) 0.583 (0.270-0.801) 0.720 (0.574-0.824) 0.719 (0.641-0.783)

5 0.733 (0.630-0.812) 0.583 (0.270-0.801) 0.720 (0.574-0.824) 0.719 (0.641-0.783)

Graft 1 0.780 (0.686-0.850) 0.750 (0.408-0.912) 0.900 (0.776-0.957) 0.815 (0.746-0.867)


dysfunction
2 0.769 (0.674-0.840) 0.750 (0.408-0.912) 0.900 (0.776-0.957) 0.808 (0.739-0.861)

3 0.746 (0.648-0.821) 0.750 (0.408-0.912) 0.880 (0.751-0.944) 0.788 (0.716-0.844)

4 0.723 (0.622-0.801) 0.667 (0.337-0.860) 0.880 (0.751-0.944) 0.767 (0.693-0.826)

5 0.710 (0.608-0.791) 0.667 (0.337-0.860) 0.854 (0.716-0.928) 0.752 (0.676-0.812)

BCAR – biopsy-confirmed acute rejection; CI – confidence interval; KM – Kaplan-Meier. The KM estimates of individual components are
presented only as a sensitivity analysis of the primary endpoint owing to potential bias due to the competing risks of occurrence of
the other primary endpoint components.

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-8 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167
ORIGINAL PAPER

1.0
+ Censored
Proportion of patients surviving without reaching the composite endpoint

0.8

0.6

0.4

0.2
De novo cohort
Conversion cohort
All patients

0.0 P value=0.603

0 1 2 3 4 5 6 7 8 9 10 11
Follow-up (years)

De novo cohort, n: 101 67 60 56 48 43 38 31 12 8 6 0


Conversion cohort, n: 62 42 38 36 33 19 13 4 1 1 0
All patients, n: 163 109 98 92 81 62 51 35 13 9 6 0

Figure 3. Kaplan-Meier plot of time from transplant to primary composite endpoint event in the full analysis cohort, de novo, and
tacrolimus conversion (combined early and late conversion) cohorts. P-value is based on time to event through 5 years.

Table 6. Kaplan-Meier estimates of patient survival stratified by population cohort (first 5 years post-transplant).

Cohort
KM rate estimate
Year
(95% CI) De novo Early conversion Late conversion Full analysis
(n=101) (n=12) (n=50) (N=163)

Patient 1 0.980 (0.923-0.995) 1.000 (1.000-1.000) 1.000 (1.000-1.000) 0.988 (0.952-0.997)


survival
2 0.941 (0.873-0.973) 0.917 (0.539-0.988) 1.000 (1.000-1.000) 0.957 (0.912-0.979)

3 0.941 (0.873-0.973) 0.917 (0.539-0.988) 1.000 (1.000-1.000) 0.957 (0.912-0.979)

4 0.921 (0.848-0.960) 0.917 (0.539-0.988) 1.000 (1.000-1.000) 0.945 (0.897-0.971)

5 0.900 (0.823-0.945) 0.917 (0.539-0.988) 0.977 (0.846-0.997) 0.925 (0.872-0.957)

CI – confidence interval; KM – Kaplan-Meier.

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-9 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
ORIGINAL PAPER Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167

Table 7. Multivariable analysis of independent variables associated with the primary composite endpoint for all patients.

Hazard ratio* Univariate hazard ratio**


Parameter P value
(95% CI) (95% CI)

Transplant center

Center 2 vs 1 0.196 (0.078-0.494) <0.001 0.372 (0.232-0.596)

Patient race/ethnicity

Black/African-American/Caribbean vs Caucasian 2.389 (0.487-11.715) 0.283 1.807 (0.435-7.503)

Middle Eastern vs Caucasian 13.519 (1.257-145.392) 0.032 1.647 (0.226-12.007)

Other vs Caucasian 6.048 (0.784-46.635) 0.084 3.760 (0.904-15.648)

Not available vs Caucasian 0.96 (0.45-2.048) 0.916 2.621 (1.641-4.187)

No. previous transplants

1 vs 0 6.592 (2.331-18.639) <0.001 2.286 (1.047-4.990)

2 vs 0 0.000 (0.000-NE) 0.994 0.000 (0.000-0.000)

Donor SCD/ECD status

ECD vs SCD 2.931 (1.554-5.526) <0.001 3.500 (2.158-5.677)

Not available vs SCD 0.394 (0.134-1.158) 0.090 0.883 (0.409-1.907)

Best eGFR***,# (mL/min/1.73 m2) 0.933 (0.894-0.974)## 0.001 0.950 (0.929-0.971)

Best serum creatinine*** (mg/dL) 0.892 (0.474-1.680) ##


0.724 2.024 (1.539-2.662)

CI – confidence interval; ECD – expanded criteria donor; eGFR – estimated glomerular filtration rate; NE – not estimable;
SCD – standard criteria donor. Proportional hazards were assumed, hazard ratio <1 indicates a reduction in hazard rate.
Center 1, Universitätsklinikum Erlangen; Center 2, Medizinische Hochschule Hannover.
* Hazard ratio generated through multivariate analysis; variables were selected using a forward selection model (P value threshold
0.05); eligible for model entry were all variables with a P value of <0.2 in univariate analysis.** Hazard ratio based on univariate Cox
regression model including the parameter as the only covariate. *** 8-12 weeks after transplant. # Estimated using modification of diet
in renal disease-4 (MDRD-4) method. ## Hazard ratio stated in relation to increases/decreases of 1 mL.

Kidney Function (crude incidence 75.0% vs 31.0-50.0% for eGFR categories


³30 mL/min/1.73 m2). The KM estimates of freedom from
In the FAC, mean serum creatinine was 1.67 mg/mL at 8-12 efficacy failure increased across the eGFR categories, with
weeks post-transplant. While all laboratory assessments were the lowest event-free rate through 5 years of 0.208 (95% CI
similar in the de novo and late conversion cohorts, metrics dif- 0.044-0.455) in the eGFR <30 mL/min/1.73 m2 category and
fered in the early conversion cohort, including decreased se- highest rate of 0.690 (95% CI 0.488-0.825) in the eGFR ³60
rum creatinine and tacrolimus trough level and increased eGFR mL/min/1.73 m2 category (Table 9).
and CKD-EPI (Table 8).

When the early conversion cohort was combined with the Discussion
late conversion cohort, mean eGFR values were similar to the
de novo cohort and remained stable through 5 years and be- This retrospective real-world analysis of KTRs from 2 study cen-
yond (Figure 5). ters in Germany was conducted to address the evidence gap in
long-term outcomes with PRT. Data were collected from trans-
Post hoc analysis of the primary composite endpoint by eGFR plant recipients receiving PRT who were also stratified into 3
at baseline (8-12 weeks post-transplant) showed that the low- sub-cohorts depending on the time from kidney transplant
est category of eGFR (<30 mL/min/1.73 m2) was associated to initiation of PRT: within the first month (de novo), within
with the highest cumulative proportion of patients reporting 6 months (early conversion from IRT), or within 3 years (late
primary composite endpoint event at 5 years post-transplant conversion from IRT). In this way, outcomes could be explored

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-10 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167
ORIGINAL PAPER

A 10.0 De novo cohort


Early conversion cohort
9.0 Late conversion cohort

8.0
Mean (SE) tacrolimus prescribed dose (mg)

7.0

6.0

5.0

4.0

3.0

2.0

1.0

0.0
0 1 2 3 4 5 6 7 8 9 10
Time from transplant (years)
De novo cohort, n: 85 83 57 69 48 68 43 62 36 58 29 57 30 40 17 14 2 8 1 5
Early conversion cohort, n: 11 10 10 11 5 8 7 7 5 6 1 2 1 1 0 0 0 0 0 1
Late conversion cohort, n: 48 46 43 43 38 42 40 36 30 26 14 17 4 5 1 3 1 – – –

B 16.0 De novo cohort


Early conversion cohort
Late conversion cohort
14.0

12.0
Mean (SE) tacrolimus through level (mg/ml)

10.0

8.0

6.0

4.0

2.0

0.0
0 1 2 3 4 5 6 7 8 9 10
Time from transplant (years)
De novo cohort, n: 92 89 83 56 69 48 68 42 63 36 59 29 57 30 40 19 16 2 9 1 6
Early conversion cohort, n: 12 11 12 10 11 5 8 7 7 5 6 1 2 1 1 0 0 0 0 0 1
Late conversion cohort, n: 47 49 44 43 42 39 42 39 36 30 26 14 17 5 5 1 3 1 – – –

Figure 4. (A) Mean prescribed tacrolimus dosages from time of transplant, stratified by de novo, early- and late-conversion cohorts.
Error bars denote standard error. (B) Mean tacrolimus trough levels from time of transplant, stratified by de novo, early- and
late-conversion cohorts. Error bars denote standard error.

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-11 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
ORIGINAL PAPER Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167

Table 8. Laboratory assessments at 8-12 weeks after transplant by population cohort.

Cohort
Parameter
De novo Early conversion Late conversion All patients
(n=92) (n=12) (n=48) (N=152)

Mean (SD) serum creatinine (mg/mL) 1.70 (0.74) 1.43 (0.64) 1.66 (0.46) 1.67 (0.65)

Mean (SD) eGFR* (mL/min/1.73 m2) 43.45 (12.37) 50.73 (19.73) 44.94 (13.04) 44.49 (13.32)

Mean (SD) CKD-EPI (mL/min/1.73 m2) 49.10 (18.52) 62.13 (24.47) 50.18 (17.47) 50.47 (18.91)

Mean (SD) tacrolimus trough level (ng/mL) 10.00 (3.04) 12.13 (5.84) 10.75 (3.12) 10.41 (3.39)

CKD-EPI – Chronic Kidney Disease Epidemiology Collaboration; eGFR – estimated glomerular filtration rate; SD – standard deviation.
* Estimated using modification of diet in renal disease-4 (MDRD-4) method.

100.0
De novo cohort
90.0 Conversion cohort

80.0

70.0
Mean (SE) eGFR (ml/min/1.73 m2)

60.0

50.0

40.0

30.0

20.0

10.0

0.0
0 1 2 3 4 5 6 7 8 9 10
Time from transplant (years)
De novo cohort, n: 92 92 89 85 54 65 45 63 34 61 27 56 20 50 21 35 14 13 0 8 0
Conversion cohort, n: 59 58 60 56 52 44 42 45 41 36 29 26 13 16 6 6 0 3 0 0 1

Figure 5. Mean (SE) eGFR values from time of transplant, stratified by de novo and combined conversion cohorts (early plus late
conversion). Patients with graft loss were included in the analysis of subsequent visits and assigned an eGFR of 0. eGFR –
estimated glomerular filtration rate; SE – standard error.
across patients with a broad spectrum of transplant-related 0.925 (95% CI 0.872-0.957), respectively, and there was no
medical histories (eg, renal impairment or rejection events pri- difference in 5-year time-to-event profiles between de novo
or to initiating PRT). The maximum period a patient could be and conversion patients. The KM estimate of median time
followed post-transplantion was 11 years and median time to primary endpoint event was 7.11 years (95% CI 3.87-NE).
between initiating PRT to discontinuation, censoring, or loss- Previous studies of IRT to PRT conversion over shorter-term ob-
to-follow-up was ~4.6 years. servation periods (1-2 years) have presented similar findings,
with no marked differences observed between early and late
For the entire population, the KM estimates of freedom from stages of conversion [28,29]. Further, a meta-analysis explor-
efficacy failure (primary composite endpoint) and overall sur- ing outcomes across 9 randomized controlled trials compar-
vival through 5 years were 0.537 (95% CI 0.455-0.612) and ing IRT or PRT in de novo KTRs found similar between-group

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-12 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167
ORIGINAL PAPER

Table 9. Kaplan-Meier estimates of primary composite endpoint stratified by baseline eGFR.

KM rate estimate (95% CI)


Year eGFR <30 ³30-<45 ³45-<60 ³60 Missing All patients
N=16 N=60 N=46 N=29 N=12 N=163
n=11 n=19 n=11 n=9 n=3 n=53
1 0.313 0.681 0.761 0.690 0.741 0.673
(0.114-0.536) (0.547-0.784) (0.610-0.860) (0.488-0.825) (0.391-0.909) (0.595-0.739)
n=11 n=23 n=14 n=9 n=3 n=60
2 0.313 0.610 0.694 0.690 0.741 0.628
(0.114-0.536) (0.473-0.721) (0.538-0.806) (0.488-0.825) (0.391-0.909) (0.549-0.698)
n=11 n=24 n=15 n=9 n=4 n=63
3 0.313 0.591 0.671 0.690 0.648 0.608
(0.114-0.536) (0.454-0.705) (0.514-0.787) (0.488-0.825) (0.310-0.852) (0.528-0.679)
n=11 n=28 n=17 n=9 n=4 n=69
4 0.313 0.516 0.623 0.690 0.648 0.567
(0.114-0.536) (0.380-0.636) (0.464-0.746) (0.488-0.825) (0.310-0.852) (0.486-0.640)
n=12 n=30 n=18 n=9 n=4 n=73
5 0.208 0.471 0.599 0.690 0.648 0.537
(0.044-0.455) (0.334-0.596) (0.440-0.726) (0.488-0.825) (0.310-0.852) (0.455-0.612)

CI – confidence interval; eGFR – estimated glomerular filtration rate in mL/min/1.73 m2; KM – Kaplan-Meier.

rates of acute rejection, graft failure, death, and adverse events The second study, a prospective multicenter 5-year follow-up
after 12 months, suggesting comparable efficacy and safe- of the ADHERE study in a European population with a demo-
ty of these 2 regimens [30]. However, the current retrospec- graphic profile that was similar to the current study in terms
tive analyses aimed to assess the longer-term outcomes in a of age, sex, and donor type, demonstrated patient survival
real-world setting in kidney transplant patients in Germany. (91.9%) and graft survival (89.3%) rates with PRT that were
Three previous studies have also reported 5-year patient and similar to those observed in the current study [22]. More re-
graft survival with PRT, but not for patients converting from cent data from the ADVANCE follow-up trial, a 5-year follow-
IRT to PRT or using a stringent composite primary endpoint up of KTRs receiving de novo PRT, reported higher patient sur-
[22,23]. The first study, an open-label, prospective, observa- vival rates (94.4%) versus the present study, but lower graft
tional, multicenter, post-marketing surveillance study in Japan survival rates (88.1%) [24]. However, the prevalence of key
[23], reported higher 5-year patient survival and graft survival factors that can influence outcomes [31,32] varied versus the
rates (96.7% and 93.2%, respectively) than those reported in present study; at most, 14.6% of patients received a kidney
the current study (92.5% and 86.2% [graft survival from sen- from a living donor in each study arm, and 65.1% of recipi-
sitivity analysis]). However, 94% of patients in the Japanese ents were male. There were also some differences in the def-
study received a kidney from a living donor (compared with inition of endpoints versus the present study.
34% in the current study), 59% of recipients were male (com-
pared with 72% in the current study), and only 7% were aged Additionally, the ADMIRAD retrospective chart analysis study
³65 years (compared with 17% in the current study). Post hoc explored long-term outcomes up to 10 years for KTRs convert-
analysis comparing the demographic characteristics and out- ing from IRT to PRT [25]. In contrast to the present study, re-
comes between Center 1 and Center 2 in our study, as well as sults for 5-year follow-up from ADMIRAD showed that the KM
previous reports [31,32], show that these factors can marked- survival rate without efficacy failure (defined as transplanta-
ly influence outcomes. Incidence of graft loss was more than tion, nephrectomy, death or return to dialysis) in ADMIRAD was
3 times greater at Center 1 versus Center 2 (25% vs 7%); of 74.1% for the PRT group and 66.7% for the IRT group and re-
which, Center 1 had a recipient population with a smaller pro- mained higher for PRT throughout follow-up (P=0.041), sug-
portion of living donors (29% vs 43%) and a larger proportion gesting a benefit for patients switching from IRT to PRT [25].
with ECD status (44% vs 11%), more patients were aged ³65
years (25% vs 10%), and with comorbidities (22% vs 7%). These In both the de novo and conversion cohorts in the present
results suggest that there may be further variables affecting study, there was a gradual reduction of mean tacrolimus daily
outcomes between the 2 centers that were not considered in dose and trough levels before doses, and trough levels became
the multivariable analysis, such as donor serum creatinine. more stable around 18 months to 2 years post-transplantation.

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-13 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
ORIGINAL PAPER Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167

Nevertheless, stable kidney function was observed in the full Conclusions


patient cohort and sub-cohorts of de novo and conversion PRT
throughout the entire period. This is consistent with previous In this retrospective study of KTRs treated with PRT in Germany,
observations where eGFR values were unaffected by reduced over 50% of patients had event-free survival through 5 years,
tacrolimus dose and trough levels after IRT to PRT conver- and overall survival was over 90%. There was no difference in
sion [33,34]. Multivariable analysis showed that donor quali- 5-year time-to-event profiles between de novo and conversion
ty, previous transplantation, and the eGFR value 8-12 weeks cohorts, and kidney function remained stable for both cohorts,
post-transplant were significantly associated with the com- with no new safety findings observed. These results support
posite endpoint for efficacy failure (P<0.001). This was also existing data demonstrating the effectiveness and safety of
confirmed in the post hoc analysis where rates of event-free long-term PRT use in KTRs.
survival 8-12 weeks post-transplant increased across the eGFR
categories from <30 mL/min/1.73 m2 to ³60 mL/min/1.73 m2. Acknowledgments

Key strengths of the current study include the long post-trans- This study was sponsored by Astellas Pharma Europe Ltd. Rhian
plantation period over which patient data extraction could Harper Owen, PhD, for Lumanity, assisted in drafting the man-
take place. However, the study also has several potential lim- uscript under the direction of the authors and provided edito-
itations: selection of patients from 2 clinical sites in Germany rial support throughout its development. S. Kadah, N.S. Heinz,
potentially introduces selection bias and may not be represen- and I. Scheffner from the Hannover Medical School (MHH), and
tative of the wider population of kidney transplant patients; M. Streubert (UK-Erlangen) also assisted with data manage-
only descriptive analyses were conducted, although this ap- ment and study organization. Medical and editorial support
proach aligns with the lack of a comparator group; and adher- was funded by Astellas Pharma, Inc.
ence with immunosuppressive therapy was not assessed, de-
spite being a known factor impacting long-term graft survival. Data Statement
There were also few patients in the early conversion cohort
and the attrition rate in the late conversion cohort resulted Researchers may request access to anonymized participant-lev-
in a low sample size by year 5, which limits the generalizabil- el data, trial-level data, and protocols from Astellas-sponsored
ity of findings for these data; one possibility for this attrition clinical trials at www.clinicalstudydatarequest.com. For the
rate may be due to graft loss experienced by patients, and Astellas criteria on data sharing see: https://clinicalstudyda-
who were then no longer in the care of the transplant center tarequest.com/Study-Sponsors/Study-Sponsors-Astellas.aspx.
(data censored). However, this graft loss would be included in
the time to composite endpoint and time to graft loss analy- Declaration of Figures’ Authenticity
ses. Despite these limitations, the current study provides cli-
nicians with real-world long-term information about the use All figures submitted have been created by the authors who
of PRT in a diverse group of recipients. confirm that the images are original with no duplication and
have not been previously published in whole or in part.

References:
1. Lamb KE, Lodhi S, Meier-Kriesche HU. Long-term renal allograft survival in 8. Sapir-Pichhadze R, Wang Y, Famure O, et al. Time-dependent variability in
the United States: A critical reappraisal. Am J Transplant. 2011;11(3):450-62 tacrolimus trough blood levels is a risk factor for late kidney transplant fail-
2. Hart A, Smith JM, Skeans MA, et al. OPTN/SRTR 2017 Annual Data Report: ure. Kidney Int. 2014;85(6):1404-11
Kidney. Am J Transplant. 2019;19:19-123 9. Rahamimov R, Tifti-Orbach H, Zingerman B, et al. Reduction of exposure
3. Gondos A, Döhler B, Brenner H, Opelz G. Kidney graft survival in Europe and to tacrolimus trough level variability is associated with better graft surviv-
the United States: Strikingly different long-term outcomes. Transplantation. al after kidney transplantation. Eur J Clin Pharmacol. 2019;75(7):951-58
2013;95:267-74 10. Gold A, Tönshoff B, Döhler B, Süsal C. Association of graft survival with ta-
4. Hariharan S, Israni AK, Danovitch G. Long-term survival after kidney trans- crolimus exposure and late intra-patient tacrolimus variability in pediatric
plantation. N Engl J Med. 2021;385(8):729-43 and young adult renal transplant recipients – an international CTS registry
analysis. Transpl Int. 2020;33(12):1681-92
5. Banas B, Krämer BK, Krüger B, et al. Long-term kidney transplant outcomes:
Role of prolonged-release tacrolimus. Transplant Proc. 2020;52(1):102-10 11. Wu MJ, Cheng CY, Chen CH, et al. Lower variability of tacrolimus trough
concentration after conversion from Prograf to Advagraf in stable kidney
6. Gaynor JJ, Ciancio G, Guerra G, et al. Graft failure due to noncompliance transplant recipients. Transplantation. 2011;92:648-52
among 628 kidney transplant recipients with long-term follow-up: A sin-
gle-center observational study. Transplantation. 2014;97:925-33 12. Stifft F, Stolk LML, Undre N, van Hooff JP, Christiaans MHL. Lower variabili-
ty in 24-hour exposure during once-daily compared to twice-daily tacrolim-
7. Borra LC, Roodnat JI, Kal JA, et al. High within-patient variability in the clear- us formulation in kidney transplantation. Transplantation. 2014;97:775-80
ance of tacrolimus is a risk factor for poor long-term outcome after kidney
transplantation. Nephrol Dial Transplant. 2010;25:2757-63 13. Tanzi MG, Undre N, Keirns J, et al. Pharmacokinetics of prolonged-release
tacrolimus and implications for use in solid organ transplant recipients.
Clin Transplant. 2016;30:901-11

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-14 [Chemical Abstracts] [Scopus]
Gwinner W. et al:
Long-term outcomes with prolonged-release tacrolimus in kidney transplantation…
© Ann Transplant, 2024; 29: e942167
ORIGINAL PAPER

14. Kuypers DRJ, Peeters PC, Sennesael JJ, et al. Improved adherence to tacro- 24. Pernin V, Glyda M, Viklický O, et al. Long-term prolonged-release tacrolim-
limus once-daily formulation in renal recipients: A randomized controlled us-based immunosuppression in de novo kidney transplant recipients: 5-y
trial using electronic monitoring. Transplantation. 2013;95(2):333-40 prospective follow-up of patients in the ADVANCE study. Transplant Direct.
15. Sellarés J, de Freitas DG, Mengel M, et al. Understanding the causes of kid- 2023;9(3):e1432
ney transplant failure: The dominant role of antibody-mediated rejection 25. Kuypers D, Weekers L, Blogg M, et al. Efficacy of prolonged-release tacrolim-
and nonadherence. Am J Transplant. 2012;12(2):388-99 us after conversion from immediate-release tacrolimus in kidney transplan-
16. Wiebe C, Gibson IW, Blydt-Hansen TD, et al. Rates and determinants of pro- tation: A retrospective analysis of long-term outcomes from the ADMIRAD
gression to graft failure in kidney allograft recipients with de novo donor- study. Transplant Direct. 2023;9(4):e1465
specific antibody. Am J Transplant. 2015;15(11):2921-30 26. von Elm E, Altman DG, Egger M, et al. The Strengthening the Reporting of
17. Rodrigo E, Segundo DS, Fernández-Fresnedo G, et al. Within-patient vari- Observational Studies in Epidemiology (STROBE) statement: Guidelines for
ability in tacrolimus blood levels predicts kidney graft loss and donor-spe- reporting observational studies. Lancet. 2007;370:1453-57
cific antibody development. Transplantation. 2016;100(11):2479-85 27. Roufosse C, Simmonds N, Clahsen-Van Groningen M, et al. A 2018 reference
18. Whalen HR, Glen JA, Harkins V, et al. High intrapatient tacrolimus variability guide to the Banff classification of renal allograft pathology. Transplantation.
is associated with worse outcomes in renal transplantation using a low-dose 2018;102:1795-814
tacrolimus immunosuppressive regime. Transplantation. 2017;101(2):430-36 28. Falconer SJ, Peagam WR, Oniscu GC. Early or late conversion from Tac-BD
19. Monchaud C, Woillard JB, Crépin S, et al. Tacrolimus exposure before and to Tac-BD in renal transplantation: When is the right time? Transplant Proc.
after a switch from twice-daily immediate-release to once-daily prolonged 2015;47(6):1741-45
release tacrolimus: The ENVARSWITCH study. Transpl Int. 2023;36:11366 29. Moal V, Grimbert P, Beauvais A, et al. A prospective, observational study of
20. Kolonko A, Słabiak-Błaż N, Pokora P, et al. Intestinal permeability in pa- conversion from immediate- to prolonged- release tacrolimus in renal trans-
tients early after kidney transplantation treated with two different formu- plant recipients in France: The OPALE study. Ann Transplant. 2019;24:517-26
lations of once-daily tacrolimus. Int J Mol Sci. 2023;24(9):8344 30. Wang TS, Huang KH, Hsueh KC, et al. Efficacy and safety of once-daily pro-
21. Caillard S, Moulin B, Buron F, et al. Advagraf®, a once-daily prolonged re- longed-release tacrolimus versus twice-daily tacrolimus in kidney trans-
lease tacrolimus formulation, in kidney transplantation: Literature review plant recipients: A meta-analysis and trial sequential analysis. J Chin Med
and guidelines from a panel of experts. Transpl Int. 2016;29:860-69 Assoc. 2023;86(9):842-49
22. Rummo O, Carmellini M, Kamar N, et al. Long-term, prolonged-release ta- 31. Auglienė R, Dalinkevičienė E, Kuzminskis V, et al. Factors influencing re-
crolimus-based immunosuppression in de novo kidney transplant recipi- nal graft survival: 7-year experience of a single center. Medicina (Kaunas).
ents: 5-year prospective follow-up of the ADHERE study patients. Transpl 2017;53(4):224-32
Int. 2020;33(2):161-73 32. Pakfetrat M, Malekmakan L, Jafari N, Sayadi M. Survival rate of renal trans-
23. Wakasugi N, Uchida H, Uno S. Safety and effectiveness of once-daily, plant and factors affecting renal transplant failure. Exp Clin Transplant.
prolonged-release tacrolimus in de novo kidney transplant recipients: 2022;20(3):265-72
5-year, multicenter postmarketing surveillance in Japan. Transplant Proc. 33. Kolonko A, Chudek J, Więcek A. Improved kidney graft function after con-
2018;50:3296-305 version from twice daily tacrolimus to a once daily prolonged-release for-
mulation. Transplant Proc. 2011;43:2950-53
34. Guirado L, Burgos D, Cantarell C, et al. Medium-term renal function in a
large cohort of stable kidney transplant recipients converted from twice-
daily to once-daily tacrolimus. Transplant Direct. 2015;1(7):e24

This work is licensed under Creative Common Attribution- Indexed in: [Science Citation Index Expanded] [Index Medicus/MEDLINE]
NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) e942167-15 [Chemical Abstracts] [Scopus]

You might also like