Open access peer-reviewed chapter

Limited Sampling Strategies to Monitoring Mycophenolic Acid Exposure in a Heterogeneous Population of Heart Transplant Recipients: A Pilot Study

Written By

Francesco Lo Re, Sandro Sponga, Jacopo Angelini, Chiara Nalli, Antonella Zucchetto, Ugolino Livi and Massimo Baraldo

Submitted: 20 December 2021 Reviewed: 27 December 2021 Published: 13 February 2022

DOI: 10.5772/intechopen.102412

From the Edited Volume

Heart Transplantation - New Insights in Therapeutic Strategies

Edited by Norihide Fukushima

Chapter metrics overview

174 Chapter Downloads

View Full Metrics

Abstract

Mycophenolate mofetil (MMF) represents a cornerstone in heart transplant (HTx) treatment. The area under the 12-hour concentration-time curve (AUC0-12h) of mycophenolic acid (MPA) -MMF’s active drug- is associated with treatment outcome. Nonetheless, therapeutic drug monitoring (TDM) of MPA AUC0-12h is impractical to assess in clinical practice and Limited Sampling Strategies (LSSs) represent a consolidated tool to estimate AUC0-12h. Two LSSs were previously generated in a selected cohort of HTx recipients treated with MMF and cyclosporine (CsA). This pilot study aimed to test these LSSs in a cohort of non-selected HTx recipients treated with MMF combined with CsA or tacrolimus (TAC). Complete PK profile was performed in 40 adults HTx recipients. MPA-AUC0-12h was estimated by two algorithms, LSS3 and LSS4, based on 3 and 4 time-points. The evaluation was made through linear regression and Bland-Altman analyses. Both LSS3 and LSS4 tended to underestimate the value of MPA-AUC0-12h (mean percentage prediction error, MPE%: −6.0%; and −4.8%, respectively). Nonetheless, high correlations (r: 0.92 and 0.94, respectively) and goodness of fit of linear regression models (R2: 0.84 and 0.88, respectively) emerged for both LSSs. A study with a wider and more homogenous sample size should be performed to support these results.

Keywords

  • heart transplantation
  • immunosuppressive treatment
  • therapeutic drug monitoring
  • treatment efficacy
  • rejection prevention

1. Introduction

Mycophenolate Mofetil MMF (CellCept; Roche, Basel, Switzerland) is a widely prescribed drug as part of maintenance immunosuppressive regimen after heart transplant (HTx) [1]. It is frequently administered in association with calcineurin inhibitors (CNIs) like cyclosporine (CsA), tacrolimus (TAC), and prednisone.

MMF is a pro-drug that, after oral administration, is rapidly hydrolyzed to its active form, mycophenolic acid (MPA), by esterases mainly in the gastrointestinal wall, blood, and liver, but also in other tissues [2]. MPA is a selective, potent and reversible inhibitor of inosine-5′-monophosphate dehydrogenase (IMPDH), a key enzyme of the de novo purine synthesis. This block causes the arrest of the proliferation of T- and B-cells [2]. In addition to this major immunosuppressive mechanism, MPA could cause the alteration of lymphocyte and monocyte recruitment, adhesion, and penetration. Furthermore, exposure to MPA could result in the apoptosis of activated human T lymphocytes, and the reduction of cytokine production. Moreover, it has been evidenced an antiproliferative effect on monocytes, fibroblasts, endothelial cells, mesangial cells, and smooth muscle cells. Nonetheless, MPA could inhibit mesangial matrix expansion, and alter the cytoskeletal organization [3, 4]. Some of these effects, including the reduction of important lymphocyte cell surface antigens expression, are independent from IMPDH inhibition [5, 6].

Generally, MMF is prescribed at a fixed dose, but there are several pharmacokinetic (PK) factors that could affect its efficacy. After MMF administration, MPA shows non-linear absorption kinetics, and a complex inter-patient and intra-patients PK variability [7], that could be attributable to MPA enterohepatic circulation (EHC), graft function, genetic factors, changes in plasma protein binding, and drug–drug interactions. MPA time to reach the plasma maximum concentration (Tmax) occurs after 1–2 hours after dosage [8].

MPA presents a higher bioavailability, ranging from 80.7–94% [8]. In blood, MPA widely binds serum albumin, from 97–99% in patients with normal renal and hepatic function. Consistently, it has been evidenced that hypoalbuminemia could increase MPA free fraction in vitro [9] and in vivo [10]. In particular, an increase of 2.2-fold of MPA free fraction emerged in vitro when MPA albumin was reduced from 41.4 g/L to 20.7 g/L, and a further increase of 41-fold when albumin was reduced to 0.07 g/L [9]. In a study including 42 adult kidney transplant recipients, a relationship between low serum albumin and an increased MPA free fraction was reported [10]. The authors identified a threshold of 31 g/L below of which MPA free fraction was considered to be significantly elevated, suggesting that the Therapeutic Drug Monitoring (TDM) of MPA free fraction could be recommended in patients with this clinical condition [10].

MPA is mainly metabolized in liver, kidney, and gastrointestinal tract by uridine 5′-diphospho-glucuronosyltransferases (UGTs). The major metabolite of MPA, 7-O-MPA-glucuronide (MPAG), is inactive but it is present in the plasma at higher concentrations than MPA. MPAG is excreted into the urine via active tubular secretion and into the bile by multi-drug resistance protein 2 (MRP-2), and at the gastrointestinal level MPAG could be de-conjugated back to MPA by gastrointestinal flora and then reabsorbed in the colon, resulting in a secondary plasma peak between 6 and 12 hours after oral administration. This may contribute to the 30–40% of MPA exposure. Severe renal impairment, liver disease, and hypoalbuminemia could affect MPA exposure [11]. The co-administration of CsA, by inhibiting the MRP-2 mainly in the gastrointestinal tract, causes a reduction of MPA EHC, resulting in an approximately 30–40% lower MPA exposure than when MMF is administered in combination with TAC [2, 8, 12]. Furthermore, it has been evidenced that CsA administration could affect MPA Clearance (Cl) [13]. Moreover, corticosteroids may reduce the exposure of MPA by inducing the expression of UGTs [8].

For these reasons, the execution of TDM could be an effective strategy to maximize the efficacy of the treatment also reduce the risk of toxicity. Several studies have suggested the importance of MPA TDM in renal and heart transplants recipients [14, 15, 16]. The best PK parameter correlating with the efficacy of treatment is represented by MPA’s area under the plasma concentration-time curve from 0 to 12 hours (MPA AUC0-12h) [11, 17] and several studies show that MPA plasma levels correlate to risk of rejection [18, 19]. The therapeutic range has been well determined in renal transplant recipients (30–60 mg × h/L) [20], and some authors suggested similar therapeutic thresholds on MPA-AUC0-12h also in HTx [21, 22].

The entire MPA AUC0-12h is difficult to calculate in clinical practice, due to its costly and laborious assessment. On the contrary, the single time-point measurement is the easiest for sampling, but it is not sufficiently predictive of patient outcome [20], taking also in consideration that MPA is characterized by >10-fold range variation in MPA AUC0-12h dose-normalized among patients undergoing heart or renal transplantation [23, 24].

Limited Sampling Strategies (LSSs) represent the most relevant assessment in solid organ transplantation for dosage individualization, that could overcome this problem [20]. LSSs are algorithm-based strategies able to predict the entire AUC0-12h without the necessity of sampling all the time-point concentrations after drug administration, but limiting the sampling to a reduced number of measurements, usually three time-points or even fewer. They can be developed by two main methods represented by multiple linear regression (MLR) or by with maximum a posteriori Bayesian estimation (MAP-BE).

MLR represents the simplest technique to develop an LSS. It requires statistical knowledge and the main strength of this approach is the adhesion to the sampling time.

On the other hand, developing an LSSs by maximum a posteriori Bayesian estimation (MAP-BE) is more complex because specialized PK modeling software knowledge is required.

From a methodological point of view, LSS should be generated on a cohort of patients (training group) and then validated in the second cohort of patients (validation group) to be used in clinical practice [25]. In the case of MLR LSSs, the relationship between the observed AUC0–12h and the estimated blood concentration-time points must be determined in the training group through linear regression, considering AUC0–12h as the dependent variable and the blood concentrations at each time point as the independent variables.

To exclude biased results, the LSS performance should be assessed in the validation group evaluating the mean prediction error or bias and the root mean squared prediction error or precision, as well as the median prediction error and the median absolute prediction error [26]. These same figures can be also calculated based on percentage prediction error, and expressed in percentages, to be more easily interpretable in the clinical contest as suggested by Baraldo et al. [25]. In both cases, the values of these parameters are inversely and proportionally linked to the LSS prediction. In the end, the correlation coefficient (r) and the coefficient of determination (R2) between the estimated and the observed AUC0-12h must be assessed.

Recently, Baraldo et al. reviewed the state of the art of MPA LSSs in HTx recipients [25]. In the last few years, the immunosuppression therapy after HTx has changed, with the massive use of TAC compared to CsA, in combination with MMF and corticosteroids.

This pilot study aimed to test, in a heterogeneous cohort of patients treated with MMF and CSA or TAC, two algorithms of LSSs previously generated by Baraldo et al. [27, 28] in a selected cohort of HTx recipients treated with MMF and CSA. These algorithms were selected due to their good performance [28] and given the hypothesis that the LSSs sampling time point schedule was able to determinate MPA AUC0-12h even when MMF was administrated combined with TAC.

If this pilot study reports positive results, the generation of new LSS in a population of HTx treated with MMF and TAC would not be required.

Advertisement

2. Methods

2.1 Study characteristics

This is a pilot observational, retrospective, cohort study. The study was performed at the University Hospital of Udine, in Italy. The study was approved by the Internal Review Board (I.R.B.) of the Commission for the Experimentation and Protection of Human Subjects of the Department of Medical Area of the University of Udine with the protocol number: 036/2020_IRB.

The study included 40 HTx recipients previously treated as per standard clinical practice with MMF and CsA or MMF and TAC, and prednisone, at the University Hospital of Udine, and routinely monitored for MPA quantification in the period starting from the 01st/01/2011 up to the 31st/12/2019. The patients included in the study were HTx recipients, aged 18 years old or more, and treated with MMF and either CsA or TAC and prednisone. Patients treated with immunosuppression drugs other than MMF, CsA and TAC, or with the absence of necessary information for the study in the clinical records or with the absence of informed consent for clinical, epidemiological research, training and study of pathologies were excluded from the study. All consecutive HTx recipients in the study period who met inclusion/exclusion criteria were included in the analysis.

All HTx recipients received a standard triple immunosuppressive therapy: MMF in combination with CsA or TAC and prednisone. The posology regimen of MMF varied from 1000 to 3500 mg/day, with a mean of 1785.5 mg/day (± 553.4). While the mean CsA dose was 3.0 mg/kg/day (± 1.3) p.o. in 2 divided doses, mean TAC dose was 0.1 mg/kg/day (±0.06). Patients treated with prokinetic drugs, resins or other drugs known to interfere with MPA PK, other than prednisone, were excluded from the analysis.

2.2 PK profiles of mycophenolate mofetil

A complete PK profile was available for the 40 HTx recipients included in the present analysis. Patients had been asked to take their usual morning dose of MMF after having a standard meal. Patients had not changed the therapeutic regimen for 30 days and had been at a steady state for MMF. Eight venous samples had been collected for the analysis of MPA plasma concentrations. For MPA assays, blood samples had been collected in EDTA tubes at 0 (pre-dose), 0.5, 1.25, 2, 4, 6, 8, and 12 hours after the morning dose. Separation of plasma was performed immediately in a centrifuge at 4°C. Plasma MPA concentration was measured using validated High Pressure Liquid Chromatography with UV Detector (HPLC/UV) method [23], that ensure to achieve an analytical precision and accuracy that fulfill the International Association of Therapeutic Drug Monitoring and Clinical Toxicology (IATDMCT) recommendations [20]. The laboratory reported the following parameters for the HPLC/UV method used for MPA quantification: limit of detection, 0.1 μg/mL; linearity, 0.1–40 μg/mL (R2: 0.9988); intrabatch imprecision (CV), 3.15%, 1.55%, and 1.76% at MPA plasma concentrations of 1.5, 5.0, 15.0 μg/mL, respectively; interbatch imprecision (CV), 3.41%, 3.21%, and 1.92% at MPA plasma concentrations of 1.5, 5.0, 15.0 μg/mL, respectively; overall inaccuracy (% Bias) of the procedure, ranged from 8.7% to 13.6%. MPA AUC0–12h had been calculated by the linear trapezoidal rule.

2.3 Algorithms evaluation

The two algorithms used for MPA AUC0-12h evaluation were the followings:

LSS3:MPAAUC012h=5.568+0.902×C1.25h+2.022×C2h+4.594×C6h
LSS4:MPAAUC012h=3.800+1.015×C1.25h+1.819×C2h+1.566×C4h+3.479×C6h

According to Sheiner and Beal, to assess the bias of the LSSs, we calculated Mean Percentage Prediction Error (MPE%) and the Median Percentage Prediction Error (MPPE%) [26]. To assess precision was calculated Root Mean Squared Percentage Prediction Error (RMSE%) and the Median Absolute Percentage Prediction Error (MAPE%) [26].

The MPE%, MPPE%, RMSE% and MAPE% were calculated as follows:

Bias:

MPE%=meanpredictedAUC012hmeasuredAUC012hmeasuredAUC012h×100%E1
MPPE%=medianpredictedAUC012hmeasuredAUC012hmeasuredAUC012h×100%E2

Imprecision:

RMSE%=meanpredictedAUC012hmeasuredAUC012hmeasuredAUC012h×100%2E3
MAPE%=medianpredictedAUC012hmeasuredAUC012hmeasuredAUC012h×100%E4

For bias, we set the limit of 15%, while for imprecision the limit was set at 20%. The percentage of estimated AUC0-12h between 75–125% of the observed AUC0-12h was also calculated.

To compare our results to an already validated algorithm, we tested one other LSS equation developed in HTx by Kaczmareck et al. [29]:

LSSKazmareck:MPAAUC012h=1.65×C0.5h+4.74×C2h

2.4 Statistical consideration

Descriptive statistical analyses were conducted for all the study variables, reporting position and variability indexes (e.g., mean and standard deviation, SD) for quantitative variables. Differences between groups were evaluated using the Fisher’s exact test for nominal variables and the Student’s T-test for quantitative variables, and considering as statistically significant a p-value <0.05.

The two methods of LSS were validated by using both linear regression and Bland–Altman analysis, as recommended by the literature [26, 30]. All the analyses were performed with Medcalc Software version 19.7.2 ® (Med-Calc Software, Ostend, Belgium®). Pearson’s linear correlation coefficient (r) was calculated using linear regression (considering the following categories for the absolute value |r|: <0.50 weak correlation, 0.50–0.80 moderate correlation; >0.80 strong correlation). The determination coefficient (R2) was also reported to assess the goodness of fit of the linear models. Bland–Altman analysis was used to evaluate the agreement between the predicted AUC0–12h and the measured AUC0–12h.

Advertisement

3. Results

3.1 Patients characteristics

The main characteristics of study patients are reported in Table 1.

TotalMMF + CsAMMF + TACp-valuea
No. pts.402812
Sex (No. males, % males)30 (75%)25 (89%)5 (42%)0.003
Age (years)56.1 ± 12.158.4 ± 10.857.5 ± 13.70.10
MMF Dose (mg/day)1785.7 ± 553.41785.7 ± 551.61791.7 ± 582.30.98
MMF Dose (mg/kg/day)24.4 ± 8.123.1 ± 6.924.3 ± 4.60.22
Post-Transpl. time (months)34.7 ± 52.545.1 ± 59.410.6 ± 140.01
BMI (Kg/m2)25.9 ± 5.426.5 ± 5.724.3 ± 4.60.21
ALT (IU/L)26.5 ± 19.528.6 ± 20.925.9 ± 15.50.24
AST (IU/L)22.9 ± 16.624.5 ± 18.619.0 ± 9.90.23
Bilirubin (mg/dL)0.9 ± 0.71.1 ± 0.70.5 ± 0.2<0.001
RBCs (x106/μL)4.1 ± 0.64.2 ± 0.73.9 ± 0.50.08
Hb (g/dL)12 ± 1.912.3 ± 1.911.4 ± 1.60.10
WBCs (x103/μL)7.7 ± 2.88.1 ± 2.86.9 ± 2.80.25
Neutro (x103/μL)5.7 ± 2.76.1 ± 2.85 ± 2.30.20
Lymph (x103/μL)1.2 ± 0.61.2 ± 0.51.1 ± 0.50.73
Mono (x103/μL)0.86 ± 1.30.9 ± 0.20.7 ± 0.20.41
Eos (x103/μL)0.09 ± 0.080.09 ± 0.10.1 ± 0.10.84
Bas (x103/μL)0.04 ± 0.030.04 ± 00.05 ± 00.74
CrCl (mL/min)b62.0 ± 26.359.7 ± 25.467.4 ± 28.70.4
GFR (ml/min/1.73m2)c59.0 ± 23.456.4 ± 24.864.4 ± 19.90.3
MPA AUC0-12h (mg × h/L)47.2 ± 24.736.4 ± 13.072.3 ± 27.60.001
MPA C0 (ug/ml)2.4 ± 2.01.6 ± 1.04.1 ± 2.6<0.001
Prednisone (mg/day)12.8 ± 9.411.9 ± 9.615.4 ± 8.80.24
Prednisone (mg/kg/day)0.2 ± 0.10.2 ± 0.10.2 ± 0.20.10
CsA Dose(mg/day)179.6 ± 75.4
CsA Dose (mg/kg/day)3.0 ± 1.3
CsA C0 (ng/mL)177.1 ± 64.9
TAC Dose (mg/day)6.0 ± 5.0
TAC Dose (mg/kg/day)0.1 ± 0.06
Tac C0 (ng/mL)10.6 ± 4.25

Table 1.

Patients baseline demographical and clinical data, overall and according to the type of treatment.

p-values of 2-sided Fisher’s exact test for nominal variables or T- test for quantitative variables.


Evaluated by Cockcroft-Gault adjusted for body weight.


Evaluated by CKD-EPI Equation.


Data are reported as mean ± standard deviation, if not otherwise specified.

AUC0-12h: Area under the plasma concentration-time curve from zero to 12 h; ALT: Alanine Aminotransferase; AST: Aspartate Aminotransferase; Bas: Basophils; BMI: Body Mass Index; C0: pre-dose measurement; CsA: Cyclosporine; CrCl: Creatinine Clearance; Eos: Eosinophils; GFR: Glomerular Filtration Rate; Hb: Hemoglobin level; Lymph: Lymphocytes; Mono: Monocytes; MMF: Mycophenolate Mofetil; MPA: Mycophenolic Acid; Neutro: Neutrophils; RBCs: Red Blood Cells; TAC: Tacrolimus; WBCs: White Blood Cells.

All patients were Caucasian and most of the analyzed patients shown normal renal and hepatic functionality. Patients treated to CsA- or TAC-based maintenance immunosuppression were comparable for most of the baseline characteristics, including age, body mass index (BMI), MMF administered dose, renal and hepatic function, except for sex, bilirubin, post transplantation time, MPA AUC0-12h and MPA C0. A number of 15 acute cell rejections occurred after a median time of 8.95 months from transplantation, especially in the patients group treated with MMF-CsA than in the MMF-TAC group (87% vs. 13%, respectively). According to the International Society for Heart and Lung Transplantation, the overall rejections were classified as follows: 8 GRADE 1R (55%), 5 GRADE 2R (33%) and 2 GRADE 3R (13%) [31]. No patients reported any episodes of diarrhea.

3.2 Method results

In the whole cohort of patients, a low tendency to underestimation of the value of MPA AUC0-12h by both LSS3 and LSS4 emerged evaluating MPE% for mean values (−6.0% and − 4.8%, respectively) and MPPE% for median values (−3.8% and − 1.1%, respectively). The precision of LSS3 and LSS4 was acceptable, by evaluating RMSE% for mean values (19.6% and 16.2%, respectively) and MAPE% for median values (13.5% and 11.0%, respectively). The percentages of MPA AUC0-12h predicted by LSS3 and LSS4 within the 25% of the MPA AUC0-12h full value was 73% and 80%, for LSS3 and LSS4, respectively.

Linear regression and Bland–Altman analyses evidenced that both LSS3 and LSS4 methods can effectively predict the values of MPA AUC0-12h. The value of r stated for both LSSs methods a strong correlation between the measured MPA AUC0-12h and the AUC0-12h predicted by both LSSs methods (r: 0.92 and 0.94 for LSS3 and LSS4, respectively). Finally, the R2 (0.84; 0.88, for LSS3 and LSS4, respectively) indicates high goodness of fit of the regression line for both methods. The results are shown in Figure 1a and b. The Bland–Altman plots (Figure 2a and b) showed that the data were arranged almost totally within the range mean +/−1.96*SD. The visual inspection of the plots does not reveal any particular pattern, thus excluding other types of bias. This was also assessed by analyzing the linear dependence of the dots in the Bland Altman plot using linear regression, reporting the following results for LSS3 and LSS4 respectively (r = 0.51 and 0.55; R2: 0.26 and 0.30). These results do not indicate linear dependence.

Figure 1.

Linear regression scatters plot of MPA AUC0-12h predicted versus MPA AUC0-12h measured, when MPA AUC0-12h predicted was calculated with LSS3 (A) and LSS4 (B) (n = 40 PK profile).

Figure 2.

Bland–Altman plots comparing MPA AUC0-12h predicted – MPA AUC0-12h measured and the average of MPA AUC0-12h predicted and MPA AUC0-12h measured, when MPA AUC0-12h predicted was calculated by LSS3 (A) and LSS4 (B) respectively (n = 40 PK profile).

A subgroup analysis was also conducted stratifying the patients for the co- treatment.

Among 28 patients treated with MMF and CsA, the bias was acceptable, evaluating MPE% for mean values (−0.5% and − 0.3%) and MPPE% for median values (2.3% and 0.7%) for LSS3 and LSS4, respectively. Analogously, the precision was acceptable evaluating RMSE% (18.6% and 14.8%) and MAPE% (12.4% and 9.7%), for LSS3 and LSS4, respectively. The percentages of MPA AUC0-12h estimated by LSS3 and LSS4 within the 25% of the MPA AUC0-12h full value were 79% and 86%, respectively.

Finally, in the sub-group of 12 patients treated with MMF and TAC, these same features were the followings: MPE% = −18.9% and − 15.3%; MPPE% = −19.9% and − 14.0%; RMSE% = 21.7% and 19.2%; MAPE% = 19.0% and 14.0%, for LLS3 and LSS4, respectively.

The percentage of MPA AUC0-12h predicted within the 25% of the measured MPA AUC0-12h full value: 58% and 67%, for LSS3 and LSS4 respectively.

Despite the very low number of patients, also the linear regression analyses executed on the two subgroups of patients evidenced good results.

In the MMF and CsA group the results were the followings: r = 0.83 and 0.89; R2 = 0.70 and 0.79, for LSS3 and LSS4 respectively; while in the MMF and TAC group these were the results: r = 0.93 and 0.93; R2 = 0.87 and 0.86, for LSS3 and LSS4 respectively. All these results are summarized on Table 2.

PopulationAlgorithmMPE (%)MPPE (%)RMSE (%)MAPE (%)% within 75–125% of full AUC0-12hR2
Overall (N = 40)LSS3−6.0−3.819.613.5730.84
LSS4−4.8−1.116.211.0800.88
MMF and CsA group (N = 28)LSS3−0.52.318.612.4790.70
LSS4−0.30.714.89.7860.79
MMF and TAC group (N = 12)LSS3−18.9−19.921.719.0580.87
LSS4−15.3−14.019.214.0670.86

Table 2.

Predictive performance of LSS3 and LSS4 in the estimation of the observed MPA AUC0-12h.

AUC0-12h: Area under the plasma concentration-time curve from zero to 12 h; CsA: Cyclosporine; LSS3: Limited Sampling Strategy based on 3 concentration sampling points; LSS4: Limited Sampling Strategy based on 4 concentration sampling points; MAPE%: Median Absolute Percentage Prediction Error; MMF: Mycophenolate Mofetil; MPA: Mycophenolic Acid; MPE%: Mean Percentage Prediction Error; MPPE%: Median Percentage Prediction Error; RMSE%: Root Mean Squared Percentage Prediction Error; R2: coefficient of determination; TAC: Tacrolimus.

The analysis of Kaczamarek LSSs applied to our patient’s data reports the following results: r = 0.70; R2 = 0.49; MPE% = 11.4% and RMSE% = 66.1% in the overall population. By applying these LSSs in the TAC subgroup of patients, we evidenced the following results: r = 0.69; R2 = 0.48; MPE% = −6.2% and RMSE% = 32.1%.

Advertisement

4. Discussion

The importance of MPA TDM for renal transplant patients is known, but its execution on HTx patients in clinical practice is still debated [17]. Specific large prospective randomized trials should be conducted, but the considerable inter- and intra-patient variability of MPA after organ transplantation suggest MPA TDM to optimize MPA exposure.

The systematic review regarding MPA TDM in HTx reported by Zuk et al. suggests that the relationship between MPA levels and the efficacy of the treatment in terms of allograft rejection in HTx patients is not defined, but LSS may be a better assessment strategy to prevent rejection than a single-time point model [32]. An LSS can be generated using two main methods: MAP-BE method and MLR analysis.

In the first case, any recorded patient sample is compared with data derived from the population PK study, and the covariates can be continually improved by updating the PK population data. The main advantage of the first approach is represented by the flexibility in the timing of the samples as recently demonstrated by Woillard et al. [22]. The main limit of this approach is represented by the employment of complex and specific software, requiring skilled professionals.

On the contrary, multiple regression analysis is simpler, but adherence to the sampling time is mandatory to apply the algorithms in clinical practice. To our knowledge, up to now, few LSSs were developed in HTx, and most of them were generated in patients treated with MMF and CsA [25]. Only three studies focused on LSSs in HTx recipients treated with MMF and TAC [29, 33, 34].

Xiang et al. [33] generated an LSS for the estimation of MMF dispersible tablets combined with TAC in 30 Chinese HTx patients. The comparison of MPA PK among MMF dispersible tablets and MMF did not show significant differences. The LSS with the best performance was the following: MPA AUC0-12h = 8.424 + 0.781 × C0.5h + 1.263 × C2h + 1.660 × C4h + 3.022 × C6h (R2 = 0.844). The performance of this LSS can be considered comparable with our algorithms and both contain the C6h sample timing point improving the MPA AUC0-12h estimation thanks to the inclusion of the typical secondary peak of MPA, minimizing the risk of MPA AUC0-12h underestimation. Nevertheless, this LSS was developed in Chinese patients so it could not properly fit the Caucasian population, although literature does not suggest this hypothesis [35]. Moreover, these LSSs were developed analyzing the plasma timing point by Liquid Chromatography with tandem mass spectrometry (LC/MS–MS), so they cannot be easily transferred in that laboratories which employ HPLC/UV methods.

Kaczmarek et al. [29] generated different LSSs in 28 HTx recipients treated with MMF and TAC. The best LSS was obtained using 4 sampling points: MPA-AUC0-12h = 1.25 × C1h + 5.29 × C4h + 2.90 × C8h + 3.61 × C10h (R2 = 0.95). The studied population is comparable to our population. Also, in this case, it can be seen that by sampling the timing point after several hours from MMF administration, a better MPA-AUC0-12h estimation can be achieved. These LSSs show an optimal performance, but it is based on a demanding sampling schedule that can be applied only on hospitalized patients, thus excluding the outpatient settings.

For this reason, authors proposed two different and more practical LSSs represented by: MPA AUC0-12h = 1.09 × C0.5 + 1.19 × C1h + 3.60 × C2h (R2 = 0.84) and MPA AUC0-12h = 1.65 × C0.5h + 4.74 × C2h (R2 = 0.75). Due to the missing data about the C1h in our population, we test the second LSS. The performance was not acceptable for the use in clinical practice as compared to our algorithms. This could be due to the absence of the C6h sampling time point, resulting in MPA AUC0-12h underestimation.

Wada et al. [34] generated an LSS in 11 Chinese HTx recipients treated MMF and TAC approximately 9 months after transplantation. In this case, the author used the same analytical method, pharmacokinetic and statistical approaches.

They generated a 3-point model LSS based on C1h, C2h and C4h: MPA AUC0-12h = 23.56 + 1.05 × C1h + 1.25 × C2h + 2.53 × C4h (R2 = 0.73), with an MPE% of 2.73%. The results of Wada’s study should be taken with caution because of the limited number of enrolled patients and the ethnic difference that could influence MPA PK.

On the other hand, Pawinski et al. proposed an accurate LSS in HTx patients treated with MMF (and CsA) [36] is based on 3 sampling time-points 2 hours after drug administration. The LSSs developed was the following: MPA AUC0-12h = 9.69 + 0.63 × C0.5h + 0.61 × C1h + 2.20 × C2h. It showed a good performance (R2 = 0.84), and for its sampling schedule it can be applied in the outpatient setting. Nevertheless, this LSS was generated on the patient in combination therapy with MMF and CsA. For this reason, this algorithm could be acceptable in patients co-treated with CsA because of its effect on reducing the typical MPA secondary peak, affecting MPA EHC [2]. Moreover, the authors developed an algorithm including the C6h blood sample. It presented a similar R2 and can be considered more predictive of the entire AUC0-12h because it can describe the typical MPA secondary peak that occurs approximately 6 to 12 hours after MMF oral dose administration, thus affecting global MPA exposure.

In our study the two evaluated LSSs reveal to be sufficiently precise and accurate for the estimation of the entire MPA AUC0-12hFigure 1. The major thesis that allows the application of these LSSs in this population is the presence of C6h that offers the opportunity to estimate MPA PK accurately in both immunosuppressive regimens, even if it is not easy to apply in the outpatient setting.

This study has several limitations: 1) the whole study group was mainly composed by men, whereas, the small subgroup of patients treated with TAC included a high percentage of women. However, it has been demonstrated that MPA PK is not influenced by sex in solid organ recipients [8, 37], even if Tornatore et al. [38] showed differences in MPA and MPAG PK related to sex among stable renal transplant recipients receiving enteric-coated mycophenolate sodium combined with TAC.; 2) in this pilot study, the sample size of the MMF and TAC group was smaller than MMF and CsA group; 3) MMF and TAC group presented a higher C0 and MPA AUC0-12h. However, exposure to MPA when MMF is in combination therapy with CsA is approximately 30–40% lower than when given in monotherapy or with TAC [8, 39]; 4) the MMF and TAC group presented a lower level of bilirubin. Bilirubin could displace MPA from albumin binding sites, affecting MPA exposure [40]. However, this effect is limited to only patients presenting hyperbilirubinemia, and could be detected only when the free drug is measured [40]; 5) TDM was not planned to be executed at the same time for all enrolled patients but it was executed by clinical decision. This can be a source of bias, because it is known that the exposition of MPA AUC0-12h could vary extensively after HTx [11]; 6) furthermore, co-medications commonly used in clinical practice could alter MPA exposure [8, 11] . However, as shown in Table 1, the major clinical parameter, including age, BMI, liver and renal function between the two treatment groups were statistically comparable.

Advertisement

5. Conclusion

In this pilot study, two LSSs resulted to be sufficiently precise and accurate to predict MPA AUC0-12h in a heterogeneous cohort of HTx patients. This study confirmed that the two LSSs, generated in HTx recipients treated with MMF and CsA could be used also in patients treated with MMF and TAC, in particular on in hospitalized patients in the first period after HTx and in outpatients with suspected toxicity or at high risk of organ rejection with considerable social, healthcare and economic advantages.

These results suggest to confirm this hypothesis in a prospective study with a wider cohort of HTx recipients, treated mainly with MMF and TAC, and with a pre-planned TDM.

Advertisement

Acknowledgments

The authors thank the healthcare staff of the Clinical Pharmacology and Toxicology Institute “University Hospital Santa Maria della Misericordia”, Udine, Italy, for the collection of blood samples and the analyses performed.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

The authors received no specific funding from any organization for this work.

Advertisement

Thanks

The authors thank Miss. Enza Pincente for the English revision of the Manuscript.

Advertisement

Nomenclature

AUC0-12hArea under the plasma concentration-time curve from zero to 12 h;
ALTAlanine Aminotransferase;
ASTAspartate Aminotransferase;
BMIBody Mass Index;
C0pre-dose measurement;
ClClearance;
CsACyclosporine;
CrClCreatinine Clearance;
EDTAEthylenediaminetetraacetic acid;
EHCenterohepatic circulation;
GFRGlomerular Filtration Rate;
HPLC/UVHigh Pressure Liquid Chromatography with UV Detector
IATDMCTInternational Association of Therapeutic Drug Monitoring and Clinical Toxicology
IMPDHinosine-5′-monophosphate dehydrogenase;
I.R.B.Internal Review Board;
LC/MS–MSLiquid Chromatography with tandem mass spectrometry;
LSSLimited Sampling Strategy;
LSS3Limited Sampling Strategy based on 3 concentration sampling point;
LSS4Limited Sampling Strategy based on 4 concentration sampling point;
MAPE%Median Absolute Percentage Predictive Error;
MPE%Mean Percentage Prediction Error;
MMFMycophenolate Mofetil;
MRP-2multi-drug resistance protein 2;
MPAMycophenolic Acid;
MPAG7-O-MPA-glucuronide;
MPPE%Median Percentage Predictive Error;
PKPharmacokinetics;
rPearson’s linear correlation coefficient;
R2coefficient of determination;
RMSE%Root Mean Squared Percentage Prediction Error
Tmaxtime to reach the plasma maximum concentration;
TACTacrolimus;
TDMTherapeutic Drug Monitoring;
UGTsuridine 5′-diphospho-glucuronosyltransferases.

References

  1. 1. Chambers DC, Cherikh WS, Harhay MO, Hayes DJ, Hsich E, Khush KK, et al. The international thoracic organ transplant registry of the International Society for Heart and Lung Transplantation: Thirty-sixth adult lung and heart-lung transplantation Report-2019; focus theme: Donor and recipient size match. The Journal of Heart and Lung Transplantation. 2019;38(10):1042-1055
  2. 2. Staatz CE, Tett SE. Pharmacology and toxicology of mycophenolate in organ transplant recipients: An update. Archives of Toxicology. 2014;88(7):1351-1389. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24792322
  3. 3. Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action. Immunopharmacology. 2000;47(2–3):85-118
  4. 4. Hackl A, Ehren R, Weber LT. Effect of mycophenolic acid in experimental, nontransplant glomerular diseases: New mechanisms beyond immune cells. Pediatric Nephrology. 2017;32(8):1315-1322
  5. 5. Gummert JF, Barten MJ, Sherwood SW, van Gelder T, Morris RE. Pharmacodynamics of immunosuppression by mycophenolic acid: Inhibition of both lymphocyte proliferation and activation correlates with pharmacokinetics. The Journal of Pharmacology and Experimental Therapeutics. 1999;291(3):1100-1112
  6. 6. Gummert JF, Barten MJ, van Gelder T, Billingham ME, Morris RE. Pharmacodynamics of mycophenolic acid in heart allograft recipients: Correlation of lymphocyte proliferation and activation with pharmacokinetics and graft histology. Transplantation. 2000;70(7):1038-1049
  7. 7. de Winter BCM, Mathot RAA, Sombogaard F, Vulto AG, van Gelder T. Nonlinear relationship between mycophenolate mofetil dose and mycophenolic acid exposure: Implications for therapeutic drug monitoring. Clinical Journal of the American Society of Nephrology. 2011;6(3):656-663
  8. 8. Staatz CE, Tett SE. Clinical pharmacokinetics and pharmacodynamics of mycophenolate in solid organ transplant recipients. Clinical Pharmacokinetics. 2007;46(1):13-58
  9. 9. Nowak I, Shaw LM. Mycophenolic acid binding to human serum albumin: Characterization and relation to pharmacodynamics. Clinical Chemistry. 1995;41(7):1011-1017
  10. 10. Atcheson BA, Taylor PJ, Kirkpatrick CMJ, Duffull SB, Mudge DW, Pillans PI, et al. Free mycophenolic acid should be monitored in renal transplant recipients with hypoalbuminemia. Therapeutic Drug Monitoring. 2004;26(3):284-286
  11. 11. Kuypers DRJ, Le Meur Y, Cantarovich M, Tredger MJ, Tett SE, Cattaneo D, et al. Consensus report on therapeutic drug monitoring of mycophenolic acid in solid organ transplantation. Clinical Journal of the American Society of Nephrology. 2010;5(2):341-358
  12. 12. Cattaneo D, Merlini S, Zenoni S, Baldelli S, Gotti E, Remuzzi G, et al. Influence of Co-medication with Sirolimus or cyclosporine on mycophenolic acid pharmacokinetics in kidney transplantation. American Journal of Transplantation. 2005;5(12):2937-2944. Available from: http://doi.wiley.com/10.1111/j.1600-6143.2005.01107.x
  13. 13. Yau W-P, Vathsala A, Lou H-X, Zhou S, Chan E. Mechanism-based enterohepatic circulation model of mycophenolic acid and its glucuronide metabolite: Assessment of impact of cyclosporine dose in Asian renal transplant patients. Journal of Clinical Pharmacology. 2009;49(6):684-699
  14. 14. van Gelder T, Hilbrands LB, Vanrenterghem Y, Weimar W, de Fijter JW, Squifflet JP, et al. A randomized double-blind, multicenter plasma concentration controlled study of the safety and efficacy of oral mycophenolate mofetil for the prevention of acute rejection after kidney transplantation. Transplantation. 1999;68(2):261-266
  15. 15. Hale MD, Nicholls AJ, Bullingham RE, Hené R, Hoitsma A, Squifflet JP, et al. The pharmacokinetic-pharmacodynamic relationship for mycophenolate mofetil in renal transplantation. Clinical Pharmacology and Therapeutics. 1998;64(6):672-683
  16. 16. DeNofrio D, Loh E, Kao A, Korecka M, Pickering FW, Craig KA, et al. Mycophenolic acid concentrations are associated with cardiac allograft rejection. The Journal of Heart and Lung Transplantation. 2000;19(11):1071-1076
  17. 17. Kiang TKL, Ensom MHH. Therapeutic drug monitoring of mycophenolate in adult solid organ transplant patients: An update. Expert Opinion on Drug Metabolism & Toxicology. 2016;12(5):545-553
  18. 18. Yamani MH, Starling RC, Goormastic M, Van Lente F, Smedira N, McCarthy P, et al. The impact of routine mycophenolate mofetil drug monitoring on the treatment of cardiac allograft rejection. Transplantation. 2000 Jun;69(11):2326-2330
  19. 19. Hesse CJ, Vantrimpont P, van Riemsdijk-van Overbeeke IC, van Gelder T, Balk AH, Weimar W. The value of routine monitoring of mycophenolic acid plasma levels after clinical heart transplantation. Transplantation Proceedings. 2001;33(3):2163-2164
  20. 20. Bergan S, Brunet M, Hesselink DA, Johnson-Davis KL, Kunicki PK, Lemaitre F, et al. Personalized therapy for mycophenolate: Consensus report by the International Association of Therapeutic Drug Monitoring and Clinical Toxicology. Therapeutic Drug Monitoring. 2021;43(2):150-200. Available from: https://journals.lww.com/10.1097/FTD.0000000000000871
  21. 21. Figurski MJ, Pawiński T, Goldberg LR, DeNofrio D, Nawrocki A, Taylor DO, et al. Pharmacokinetic monitoring of mycophenolic acid in heart transplant patients: Correlation the side-effects and rejections with pharmacokinetic parameters. Annals of Transplantation. 2012;17(1):68-78
  22. 22. Woillard J-B, Saint-Marcoux F, Monchaud C, Youdarène R, Pouche L, Marquet P. Mycophenolic mofetil optimized pharmacokinetic modelling, and exposure-effect associations in adult heart transplant recipients. Pharmacological Research. 2015;99:308-315. Available from: https://www.sciencedirect.com/science/article/pii/S1043661815001450
  23. 23. Shaw LM, Korecka M, van Breeman R, Nowak I, Brayman KL. Analysis, pharmacokinetics and therapeutic drug monitoring of mycophenolic acid. Clinical Biochemistry. 1998;31(5):323-328
  24. 24. Shaw LM, Kaplan B, DeNofrio D, Korecka M, Brayman KL. Pharmacokinetics and concentration-control investigations of mycophenolic acid in adults after transplantation. Therapeutic Drug Monitoring. 2000;22(1):14-19
  25. 25. Baraldo M, Sponga S, Livi U. Therapeutic drug monitoring of Micophenolate Mofetil in cardiac transplant patients by limited sampling strategy: An update. In: Rescigno G, Firstenberg MS, editors. Topics in Heart Failure Management [Internet]. Rijeka: IntechOpen; 2018. Available from: https://www.intechopen.com/books/topics-in-heart-failure-management/therapeutic-drug-monitoring-of-micophenolate-mofetil-in-cardiac-transplant-patients-by-limited-sampl
  26. 26. Sheiner LB, Beal SL. Some suggestions for measuring predictive performance. Journal of Pharmacokinetics and Biopharmaceutics. 1981;9(4):503-512
  27. 27. Baraldo M, Isola M, Feruglio MT, Francesconi A, Franceschi L, Tursi V, et al. Therapeutic mycophenolic acid monitoring by means of limited sampling strategy in orthotopic heart transplant patients. Transplantation Proceedings. 2005;37(5):2240-2243
  28. 28. Baraldo M, Cojutti PG, Isola M, Feruglio MT, Tursi V, Livi U, et al. Validation of limited sampling strategy for estimation of mycophenolic acid exposure during the first year after heart transplantation. Transplantation Proceedings. 2009;41(10):4277-4284
  29. 29. Kaczmarek I, Bigdeli AK, Vogeser M, Mueller T, Beiras-Fernandez A, Kaczmarek P, et al. Defining algorithms for efficient therapeutic drug monitoring of mycophenolate mofetil in heart transplant recipients. Therapeutic Drug Monitoring. 2008;30(4):419–427
  30. 30. Bland JM, Altman DG. Statistical methods for assessing agreement between two methods of clinical measurement. Lancet (London, England). 1986;1(8476):307-310
  31. 31. Billingham M, Kobashigawa JA. The revised ISHLT heart biopsy grading scale. The Journal of Heart and Lung Transplantation: the Official Publication of the International Society for Heart Transplantation. 2005;24:1709
  32. 32. Zuk DM, Pearson GJ. Monitoring of mycophenolate mofetil in orthotopic heart transplant recipients--a systematic review. Transplantation Reviews (Orlando, Fla.). 2009;23(3):171-177
  33. 33. Xiang H, Zhou H, Zhang J, Sun Y, Wang Y, Han Y, et al. Limited sampling strategy for estimation of mycophenolic acid exposure in adult chinese heart transplant recipients. Frontiers in Pharmacology. 2021;12:652333
  34. 34. Wada K, Takada M, Kotake T, Ochi H, Morishita H, Komamura K, et al. Limited sampling strategy for mycophenolic acid in Japanese heart transplant recipients. Circulation Journal. 2007;71(7):1022-1028
  35. 35. Ling J, Shi J, Jiang Q, Jiao Z. Population pharmacokinetics of mycophenolic acid and its main glucuronide metabolite: A comparison between healthy Chinese and Caucasian subjects receiving mycophenolate mofetil. European Journal of Clinical Pharmacology. 2015;71(1):95-106
  36. 36. Pawinski T, Kunicki PK, Sobieszczanska-Malek M, Gralak B, Szlaska I. A limited sampling strategy for estimating mycophenolic acid area under the curve in adult heart transplant patients treated with concomitant cyclosporine. Journal of Clinical Pharmacy and Therapeutics. 2009;34(1):89-101
  37. 37. Pescovitz MD, Guasch A, Gaston R, Rajagopalan P, Tomlanovich S, Weinstein S, et al. Equivalent pharmacokinetics of mycophenolate Mofetil in African-American and Caucasian male and female stable renal allograft recipients. American Journal of Transplantation. 2003;3(12):1581-1586. DOI: 10.1046/j.1600-6135.2003.00243.x
  38. 38. Tornatore KM, Meaney CJ, Wilding GE, Chang SS, Gundroo A, Cooper LM, et al. Influence of sex and race on mycophenolic acid pharmacokinetics in stable African American and Caucasian renal transplant recipients. Clinical Pharmacokinetics. 2015;54(4):423-434
  39. 39. van Gelder T, Klupp J, Barten MJ, Christians U, Morris RE. Comparison of the effects of tacrolimus and cyclosporine on the pharmacokinetics of mycophenolic acid. Therapeutic Drug Monitoring. 2001;23(2):119-128
  40. 40. Shaw LM, Figurski M, Milone MC, Trofe J, Bloom RD. Therapeutic drug monitoring of mycophenolic acid. Clinical Journal of the American Society of Nephrology. 2007;2(5):1062 LP-1061072. Available from: http://cjasn.asnjournals.org/content/2/5/1062.abstract

Written By

Francesco Lo Re, Sandro Sponga, Jacopo Angelini, Chiara Nalli, Antonella Zucchetto, Ugolino Livi and Massimo Baraldo

Submitted: 20 December 2021 Reviewed: 27 December 2021 Published: 13 February 2022