• No results found

University of Groningen Towards personalized cardiovascular risk management in renal transplant recipients de Vries, Laura Victorine

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Towards personalized cardiovascular risk management in renal transplant recipients de Vries, Laura Victorine"

Copied!
25
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Towards personalized cardiovascular risk management in renal transplant recipients

de Vries, Laura Victorine

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2018

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

de Vries, L. V. (2018). Towards personalized cardiovascular risk management in renal transplant recipients.

Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 7

(3)
(4)

7

SUMMARY AND GENERAL DISCUSSION

Prelude

Cardiovascular risk is greatly increased in renal transplant recipients (RTR), which likely is due to an interaction of traditional and transplantation-related cardiovascular risk factors. Many RTR already have several traditional cardiovascular risk factors before transplantation, which are only partially remitted following successful transplanta-tion, and in fact, are more prevalent, more severe or less responsive to treatment than in non-transplanted patients. In addition, new transplantation-related risk factors emerge, such as remaining subnormal kidney function and the use of immunosuppres-sive drugs. Moreover, within the current immunosuppresimmunosuppres-sive regimens, corticosteroids are still indispensable and dosed by a more or less ‘one-size-fits-all’ empirical schedule and therefore may result in higher administered doses than strictly necessary for an adequate therapeutic effect, which potentially adds to the already increased cardiovas-cular risk in these patients. Therefore, we are in need of comprehensive strategies to reduce the increased cardiovascular risk in RTR, ideally addressing both traditional and transplantation-related risk factors, and targeting the specific risk profile in this patient category rather than extrapolating from guidelines based on studies in other popula-tions. In addition, we are in need of biomarkers that allow for better personalization of treatment. In this thesis, we focused on potential strategies to reduce hyperten-sion, which is the most prevalent cardiovascular risk factor in RTR, specifically focusing on dietary sodium restriction (Chapter 2), the renin-angiotensin-aldosterone-system (RAAS), and their interaction (Chapter 3). In addition, we aimed to find biomarkers that allow for personalization of corticosteroid therapy, specifically focusing on path-ways of endogenous cortisol synthesis (Chapters 4 and 5) and chronic inflammation (Chapter 6).

Treating post-transplantation hypertension: a role for dietary sodium restriction?

Hypertension is the most prevalent cardiovascular risk factor in RTR. Up to 90% of RTR have high blood pressure or are treated with antihypertensive drugs.1-3 However,

antihypertensive treatment in RTR often remains suboptimal, with the vast majority of patients not meeting recommendations of a blood pressure below 130/80 mmHg, or even below 140/90 mmHg.1 Dietary sodium restriction is an important treatment

strategy in chronic kidney disease (CKD), as recommended in guidelines for cardiovas-cular risk management in CKD.4 However, prior to our study, sodium restriction has

not been investigated in an appropriately designed study in RTR. In our study, which is described in Chapter 2, we aimed to investigate the effects of dietary sodium restric-tion on blood pressure and albuminuria in stable RTR, in a two-center randomized cross-over clinical trial. To this end, we randomly assigned 23 stable RTR to treatment

(5)

with either a 6-week regular sodium diet (target: 150 mmol of sodium per day, equaling 9 grams of salt per day) or a 6-week low sodium diet (target: 50 mmol of sodium per day, equaling 3 grams of salt per day), after which they were crossed-over to the other treatment arm. Adherence to the sodium diet was monitored by measuring urinary sodium excretion in 24h urine samples, and adequate adherence was achieved in 86% of patients. We found that dietary sodium restriction reduced both systolic and diastolic blood pressure and, when adherence with the diet was adequate, also albu-minuria, without materially affecting kidney function.

As already mentioned, our study is the first to investigate the effects of dietary sodium restriction in RTR, using a randomized trial design. However, it should be noted that this study in RTR is only the most recent in line of a long tradition of clinical trials on dietary sodium restriction in patients with non-diabetic and diabetic CKD by our5-10 and other groups.11 All these previous studies consistently showed that

dietary sodium restriction significantly reduces blood pressure and albuminuria, and potentiates the efficacy of treatment with RAAS blockers. In addition, in CKD patients, adding dietary sodium restriction to treatment with RAAS blockade was shown to be as effective as adding hydrochlorothiazide7,9 and even more effective

than treatment with a combination of two RAAS blocking agents.8 Moreover, several

studies show that low sodium intake is associated with much better long-term renal and cardiovascular outcomes in CKD patients.12,13 In contrast, high sodium intake has

been shown to blunt the antiproteinuric effects of RAAS blockade in CKD patients,14

and also recently in RTR.15

We found that dietary sodium restriction reduced systolic blood pressure by 11 mmHg and diastolic blood pressure by 7 mmHg (Chapter 2). Compared to the 4 to 6 mmHg reduction in mean arterial pressure by dietary sodium restriction in CKD patients,5-10

blood pressure reduction in our study was quite impressive. Indeed, blood pressure is thought to be very sodium sensitive in RTR. This was also illustrated by a recent study in 660 Dutch RTR that showed an independent association of sodium intake with blood pressure.16 Contributing factors to sodium sensitivity of blood pressure may include

factors which are also present in CKD patients, such as prevalent obesity,17,18 reduced

kidney function, and proteinuria.7,19 In addition, treatment with immunosuppressive

drugs, such as corticosteroids and/or calcineurin inhibitors (CNIs), may render blood pressure even more sodium sensitive.20 More specifically, corticosteroids are known

to activate the mineralocorticoid receptor, resulting in increased water and sodium retention.21,22 In addition, CNIs increase sodium sensitivity by activation of the

(6)

7

Taking this into consideration, it may seem very surprising that dietary sodium restric-tion is not applied more systematically and effectively in clinical practice in RTR yet. This may be related to the overall lack of adequate lifestyle management relative to phar-macological management in clinical medicine, but there may also be transplant-specific barriers. One of these could be the reluctance of transplant physicians to apply antihy-pertensive treatment strategies which may give an initial reduction in kidney function,25

such as the use of RAAS blockers, (thiazide) diuretics, and dietary sodium restriction, or a combination thereof. Obviously, a treatment-related reduction in kidney function is less desirable shortly after transplantation, because it can confound the assessment of possible rejection. However, in patients who have stable transplant function but have inadequately treated hypertension and/or proteinuria with a combination of antihy-pertensive drugs, it may be a different story. Especially, since the reduction in kidney function is often temporary and not related to acute kidney injury. Moreover, a slight reduction in kidney function following treatment with RAAS blockade has been shown to predict better kidney outcomes in the long-term in CKD patients, possibly because it indicates amelioration of glomerular hypertension.26,27 Despite the rigorous reduction

in sodium intake, serum creatinine rose only slightly in our study. Therefore, our study demonstrates that sodium restriction could be feasible in RTR, without compromising allograft function too severely.

Of note, choosing the most suitable antihypertensive treatment strategy in RTR remains a challenge to date, because few studies have directly compared antihy-pertensive agents in this population.25,28 The majority of studies compared a single

antihypertensive drug, mostly a calcium channel blocker or angiotensin-converting enzyme inhibitor, with no treatment or placebo,28,29 while very often treatment with two

or more antihypertensive drugs is required. In addition, most studies only assessed intermediate end points, whereas the choice of antihypertensive therapy should ide-ally be guided by hard end points, like cardiovascular events or allograft failure. More importantly, there are no large prospective studies investigating effects of lifestyle interventions, such as dietary sodium restriction, or multimodal lifestyle approaches in RTR. Such studies would be highly valuable, and remain sorely awaited. Meanwhile, valuable information can be obtained from well-documented observational cohorts, such as the TransplantLines cohort in our center, and from short-term interventions on intermediate end-points. Interestingly, when intermediate end-points are available, such as is the case for hypertension, or insulin resistance, rotation design studies allow to identify personalized treatment strategies.30 However, even in the general

popu-lation, despite long-term availability of data and strategies to support personalized approaches in antihypertensive treatment, so far, the concept has not systematically been elaborated, and thus is considered to be still in its infancy.31

(7)

Another potential obstacle to applying dietary sodium restriction in clinical practice may be adherence to the sodium restricted diet. Current sodium intake of stable RTR in the Netherlands is about 150-200 mmol (equaling about 9-12 grams of salt) per day,1,16

which is comparable to sodium intake in the general population, and by far exceeds the recommended intake of less than 100 mmol (or 6 grams of salt) per day.32,33 We

and others showed that an 80- to 100-mmol reduction in sodium intake is feasible in a regular nephrology outpatient setting, at least for the duration of the studies (Chapter 2).7,8,10 However, when patients participated in these studies, they received

intensive, individualized, dietary counseling by study physicians and/or dieticians and adherence to the study diet was checked by interim urinary sodium analysis. This most probably led to increased dietary adherence. In addition, there is mounting evidence that persistent lifestyle alterations necessitate a dedicated behavioral approach. Such strategies are not included in routine clinical care yet, but have recently been studied in e.g. the Sodium Burden Lowered by Lifestyle Intervention: Self-Management and E-Health Technology [SUBLIME] Study.34 This study in CKD patients and RTR showed

that the addition of a program using e-health technology and self-management to regular dietary counseling was highly valued by the patients, and also the feedback on actual dietary intake of sodium by the on-line tool, as well as the 24h urine excretion of sodium was considered very useful. In addition, it resulted in a sustained reduction in dietary sodium intake and blood pressure. Therefore, to really achieve a persistent reduction in dietary sodium intake in RTR, we need comprehensive strategies, which encompass individualized dietary counseling and include a behavioral approach, including feedback on actual dietary intake.

Sodium and aldosterone: evil twins in action

In Chapter 3 we reviewed the role of the mineralocorticoid hormone aldosterone in the pathophysiology of cardiovascular and renal damage. Aldosterone is classically known for its role in water and sodium retention via activation of the mineralocorticoid receptor (MR), and therefore for its role in inducing hypertension. We describe that its actions go far beyond these classical effects, via mechanisms involving oxidative stress, inflammation, and fibrosis, which occur both in the heart and kidneys. Furthermore, we provide evidence that detrimental effects of aldosterone are likely only observed in states of primary increase in aldosterone concentrations, rather than states in which increased aldosterone concentrations are secondary to volume depletion. For example, in patients with resistant hypertension, the effects of high sodium intake on proteinuria are most pronounced in patients with the highest aldosterone.35 In

con-trast, in case of hyperaldosteronism secondary to volume depletion, such as routine low-sodium intake in Yanomami Indians or Gitelman or Bartter syndrome with renal sodium loss, hypertension and cardiovascular damage are absent.36,37 Taken together,

(8)

7

these data suggest that deleterious effects of aldosterone are most pronounced when its serum concentration is inappropriately high for the prevailing sodium status. Therefore, MR blockade may have great potential to halt the progressive end organ damage as observed during current treatments, especially in combination with dietary sodium restriction. Indeed, the effectiveness of MR blockade in heart failure has already been proven in large randomized clinical trials.38,39 In addition, numerous small-scale

clinical studies have demonstrated increased treatment efficacy by the addition of MR blockade to RAAS blockade in patients with non-diabetic40-42 and diabetic CKD.43,44

How-ever, long-term data on the efficacy and safety of MR blockade in preventing dialysis and/or cardiovascular endpoints in CKD are still lacking. Moreover, there are currently no studies available in RTR, which investigate the effects of MR blockade on blood pressure and/or albuminuria, or long-term patient and allograft survival. Therefore, it would be of great interest to further explore whether treatment with MR blockade, possibly in combination with dietary sodium restriction, may also be beneficial in RTR.

From mineralocorticoids to glucocorticoids: monitoring prednisolone therapy

Chronic prednisolone treatment after kidney transplantation is associated with numerous side effects, including hypertension and impaired insulin sensitivity, which contribute to increased cardiovascular risk in RTR.45,46 In addition, prednisolone dosing

regimens remain empiric to date, usually with fixed doses, independent of either body size and/or steroid sensitivity.47 The main reason is that there is currently no way to

guide intensity of treatment, potentially leading to even more unwanted side effects. Chronic prednisolone treatment is also known to suppress endogenous cortisol production, by negative feedback inhibition of the hypothalamus-pituitary-adrenal (HPA)-axis.48-50 In addition, treatment with exogenous corticosteroids has been

sug-gested to alter systemic cortisol exposure by interfering in the enzymes that activate and inactivate cortisol, i.e. the 11-beta hydroxysteroid dehydrogenases (11β-HSDs). 51-53 We hypothesized that the degree to which prednisolone affects HPA axis and/or

11β-HSD enzyme activity might reflect the pharmacological effects of prednisolone, and might, therefore, be related to prednisolone-related metabolic side effects and long-term outcome after kidney transplantation.

In Chapter 4 we investigated whether HPA axis function, as measured by 24h urinary free cortisol excretion, is altered in prednisolone-treated RTR, and whether the degree of HPA axis suppression is related to prednisolone-related metabolic side effects and the metabolic syndrome. In a large cohort of 563 stable RTR, we found that urinary free cortisol excretion was markedly decreased compared to known values for the general population. Interestingly, there was considerable inter-individual variation in urinary

(9)

free cortisol excretion, even in patients treated with the same prednisolone dose. Moreover, we found that a higher degree of HPA axis suppression by prednisolone, as reflected by lower 24h urinary free cortisol excretion, was associated with increased prevalence of the metabolic syndrome and its individual components (obesity, dyslip-idemia, hypertension, and diabetes).

In Chapter 5 we went one step further into cortisol metabolism, and investigated not only HPA axis activity, but also 11β-HSD enzyme activities in 693 stable pred-nisolone-treated RTR and compared these to 275 healthy controls. In addition, we investigated whether altered HPA axis and 11β-HSD enzyme activities were related to long-term outcome after kidney transplantation. To this end, we measured 24h uri-nary excretion of total cortisol, but also of its inactive counterpart cortisone, and their metabolites tetrahydrocortisol (THF), allo-tetrahydrocortisol (alloTHF), and tetrahydro-cortisone (THE). Urinary total cortisol excretion and summated excretion of cortisol, cortisone and their metabolites were used as estimates of HPA axis activity. Urinary (THF+alloTHF)/THE and cortisol/cortisone ratios were used as estimates for 11β-HSD enzyme activities. We found that urinary cortisol excretion and summated cortisol and metabolite excretion were significantly lower in RTR compared with healthy controls, whereas urinary (THF+alloTHF)/THE and cortisol/cortisone ratios were significantly higher. Again, there was considerable variation in these parameters in patients treated with the same prednisolone dose. In addition, we found that a higher degree of HPA axis suppression, as reflected by lower urinary cortisol and metabolite excretion, and also higher (THF+alloTHF)/THE and cortisol/cortisone ratios, were associated with increased risk of mortality, independent of potential confounders.

HPA axis activity in this thesis was estimated by 24h urinary excretion of free cortisol in

Chapter 4, and by 24h urinary excretion of both total cortisol and summated urinary excretion of cortisol and its metabolites in Chapter 5. We chose to measure urinary excretion of free cortisol in Chapter 4, because this has often been used as a proxy for HPA axis activity and is considered to be the biologically active fraction.54 However,

endogenous glucocorticoids are present in urine largely as conjugated derivatives (95% as glucuronides, and 3-4% as sulfates).55,56 Therefore, we think that measuring total

cortisol, as we did in Chapter 5, instead of free urinary cortisol could potentially provide a more reliable reflection of total cortisol production. In addition, cortisol is mainly secreted in the urine in the form of its metabolites.57 As a result, the sum of urinary

THF, alloTHF, and THE encompasses about 50% of total endogenous glucocorticoid secretion.55,57 Thus, summation of urinary cortisol and its metabolites, as we

addition-ally did in Chapter 5, might even better reflect HPA axis activity than cortisol alone. We found that HPA axis activity was suppressed in prednisolone-treated RTR compared

(10)

7

to subjects from the general population. This is in line with previous studies showing that chronic prednisolone treatment suppresses HPA axis activity after kidney trans-plantation.48-50 However, substantial residual endogenous cortisol synthesis was still

present in the majority of patients, despite treatment with prednisolone. Daily cortisol production rate in healthy subjects has been estimated to be approximately 6-10 mg/ m2/day,58,59 which is equivalent to 3-5 mg of prednisolone per day. When the

equiv-alent exogenous glucocorticoid dose is higher than endogenous cortisol production, endogenous cortisol production is usually suppressed. Thus, theoretically, when a patient is chronically treated with a daily dose of prednisolone greater than 5 mg/ day, and certainly greater than 7.5 mg/day, one might expect that HPA axis activity is entirely suppressed, with no residual endogenous cortisol synthesis present. How-ever, our data, showing considerable endogenous cortisol synthesis in patients with daily prednisolone doses of 7.5 mg and higher, indicate that this is not what actually happens. Indeed, biological half-life of prednisolone is only 2-4 hours in stable outpa-tient RTR.60-64 Thus, one daily dose of prednisolone, like conventionally prescribed in

our center and many other transplant centers, may not fully cover the normal diurnal pattern of endogenous cortisol requirement and thus may leave room for endogenous production.

Interestingly, we found that there was considerable inter-individual variation in HPA axis activity, even in patients treated with the same prednisolone dose (Chapters 4 and 5). At first this seemed a surprising result, but when we examined both prednisolone pharmacokinetics and individual glucocorticoid sensitivity in more detail, this seemed not so surprising after all. First, from literature it is known that there is a greater than 3-fold variability in dose-adjusted prednisolone exposure in solid organ transplant recipients.47 Many factors contribute to this variability, of which the most well-studied

are patient age, sex, and kidney function.47 Female sex has consistently been

associ-ated with increased prednisolone exposure, with a typical female having more than half the clearance of a typical male.64 In addition, prednisolone clearance is reduced

in patients with reduced kidney function.65-68 Finally, in elderly transplant recipients,

despite reduced metabolism of prednisolone, endogenous cortisol production has also been reported to be higher, suggesting less suppression of endogenous cortisol production by prednisolone.69 In line with this, we found higher age, male sex, and

better kidney function to be independently associated with higher urinary cortisol and metabolite excretion (Chapter 5). Still, a greater understanding of the influence of various patient factors on prednisolone exposure in RTR is needed.

Moreover, the high inter-individual variation in HPA axis activity under prednisolone treatment we found, might also be related to the degree of individual sensitivity to

(11)

(exogenous and/or endogenous) glucocorticoids. Glucocorticoids are involved in count-less metabolic and immune-modulatory processes. Therefore, it comes to no surprise that their regulation is extremely complex, involving a myriad of regulatory steps, which are often tissue and cell-specific.70 Individual variation in any of these steps, therefore,

contributes to large variability in individual glucocorticoid sensitivity, and also to vari-ability in glucocorticoid sensitivity of different tissues. It is beyond the scope of this discussion to go in great detail into this subject. In brief, glucocorticoids bind to the glucocorticoid receptor (GR) to exert their effects. Availability of glucocorticoids for the GR depends on the degree to which they are free or bound to cortisol binding globulin (CBG) or albumin in plasma.71 In addition, glucocorticoid availability is regulated by

the degree to which glucocorticoids are inactivated to their inactive counterparts by the 11β-HSD2 enzyme and regenerated back by 11β-HSD1.53,72 Therefore, individual

variation in CBG and 11β-HSD enzyme activity could contribute to individual gluco-corticoid sensitivity. In addition, the number and affinity of available binding sites for glucocorticoids to the GR vary within the healthy population.73 This is caused by, among

others, alternative splicing of the gene coding for the GR, giving rise to GR isoforms,74

and genetic polymorphisms in the GR.75 Thus, inter-individual variation in innate

gluco-corticoid sensitivity, in combination with differences in prednisolone exposure, could potentially lead to considerable variation in HPA axis activity in patients treated with the same prednisolone dose. Therefore, it is conceivable that under a standard once-daily dosing regimen with relatively low dosages, there can be incomplete suppression of the HPA axis and that there is considerable variation in the extent to which the HPA axis is suppressed.

We found that (THF+alloTHF)/THE and cortisol/cortisone ratios were increased in RTR compared to healthy controls (Chapter 5). Higher urinary (THF+alloTHF)/THE and cortisol/cortisone ratios suggest that cortisol balance as maintained by 11β-HSD enzymes has shifted towards cortisol production rather than metabolism. Since 11β-HSD2 metabolizes cortisol to cortisone and 11β-HSD1 regenerates cortisol back from cortisone, a shift towards cortisol production could either be attributable to a relative decrease in enzymatic activity of 11β-HSD2 or relative increase in enzymatic activity of 11β-HSD1. The 11β-HSD2 enzyme is mainly localized in mineralocorticoid sensitive tissues such as the kidney, where it protects the MR from cortisol. Previous stud-ies showed that expression of 11β-HSD2 is reduced in kidney failure.76-79 We found

a strong inverse association of (THF+alloTHF)/THE and cortisol/cortisone ratios with kidney function, so this could also be the case in our study. However, it has recently been suggested that exogenous steroids, such as prednisolone, could also influence peripheral cortisol balance by induction of 11β-HSD1, thereby creating a feed-forward mechanism enhancing systemic cortisol exposure.51-53 Taking this into consideration,

(12)

7

maybe not only the degree of HPA axis suppression, as estimated by 24h urinary corti-sol and metabolite excretion, but also (THF+alloTHF)/THE and corticorti-sol/cortisone ratios in our study could be a reflection of the pharmacological effects of prednisolone. This is supported by the very strong inverse, kidney function independent, association of uri-nary summated cortisol and metabolite excretion with (THF+alloTHF)/THE ratio, which we found in our study. Moreover, both decreased 24h urinary cortisol and metabolite excretion and increased (THF+alloTHF)/THE and ratio were associated with increased risk of all-cause mortality, and mortality of cardiovascular and infectious causes, which associations were also all independent of kidney function.

Although individual glucocorticoid sensitivity is more and more acknowledged as important determinant of potential success and toxicity of corticosteroid treatment,70,75

there are few to no clinical read-out parameters by which it can be assessed. We chose to measure 24h urinary excretion of cortisol, cortisone, and their metabolites to measure prednisolone-related effects on the HPA axis. In addition, we measured (THF+alloTHF)/THE and cortisol/cortisone ratio to estimate systemic 11β-HSD enzyme activities. We realize that these are very crude measures to assess something as del-icate is glucocorticoid sensitivity, but they are – in our opinion – the best available clinical read-out parameters available yet. Indeed, we could have measured pred-nisolone exposure by means of pharmacokinetic models. Nevertheless, measuring exposure to prednisolone would not have given us information on its effects. Mea-surement of urinary excretion of cortisol and/or its metabolites has the advantage of being non-invasive, relatively easy, and more feasible for large cohort studies and future application in patient care. In addition, it is relatively unaffected by the circadian rhythm of cortisol or by varying plasma protein binding capacities compared to serum cortisol. Thus, although we made a first step towards measuring individual sensitivity to the effects of prednisolone, it would be of great interest to explore alternative ways to measure individual glucocorticoid sensitivity to personalize prednisolone therapy. One of the main drawbacks of the studies presented in Chapters 4 and 5, is that they are observational in nature. Observational studies can only establish associations between observations and outcome variables, but cannot establish whether the asso-ciations identified represent cause-and-effect relationships, leaving room for potential confounding. Although we adjusted for several potential confounding factors, such as age and sex, daily prednisolone dose, kidney function and body composition, residual confounding cannot be excluded. In addition, medication intake was not monitored in these studies. Therefore, we did not have information on adherence to treatment with corticosteroids or other immunosuppressive drugs, or the time of the day patients took these drugs. This is especially important since prednisolone half-life is typically

(13)

2-4 hours.60-64 In addition, the rate of non-adherence to immunosuppressive

medica-tion has been reported to range from 15% to even 45% in RTR.80,81 Due to the lack of

information on adherence to prednisolone intake, this is something we could not take into account and could have affected our results. Moreover, all but two patients in our studies were treated with prednisolone as part of their maintenance immunosuppres-sive regimen, which makes it difficult to dissect whether alterations in HPA axis and 11β-HSD enzyme activities were related to treatment with prednisolone or are innate to the situation of kidney transplantation itself. Thus, to address all these questions, we are currently investigating cortisol metabolism in a post-hoc analysis of a randomized clinical trial in which intake of immunosuppressive medication was closely monitored, including intake of prednisolone, and which also had a prednisolone-free arm.

The tryptophan-kynurenine pathway: friend or foe in transplantation?

A major effect of corticosteroids is to suppress inflammation, not only locally in trans-planted organs, but also systemically, in the organism hosting the transtrans-planted organ. In RTR, systemic inflammation is associated with reduced long-term patient82,83 and

allograft survival.84,85 An interesting pathway that is tightly linked to systemic

inflam-mation and corticosteroid exposure is the kynurenine pathway, which is the major metabolic pathway of the essential amino-acid tryptophan. Under inflammatory condi-tions, tryptophan is metabolized by the enzyme indoleamine 2,3-dioxygenase (IDO) to kynurenine, which is subsequently metabolized by the enzyme kynurenine 3-monooxy-genase (KMO) to cytotoxic 3-hydroxykynurenine.86-88 Therefore, in Chapter 6, we aimed

to investigate whether activation of the tryptophan-kynurenine pathway is associated with systemic inflammation in stable RTR, and whether it is also associated with long-term patient and allograft survival.

To this end, we measured serum and urinary tryptophan, kynurenine, and 3-hydroxyky-nurenine in a cohort of 561 stable RTR. In addition, IDO enzyme activity was estimated by the kynurenine-to-tryptophan ratio and KMO enzyme activity was estimated by the 3-hydroxykynurenine-to-kynurenine ratio. We found that serum kynurenine and particularly cytotoxic serum 3-hydroxykynurenine, and also serum kynurenine-to-tryp-tophan and 3-hydroxykynurenine-to-kynurenine ratios, were strongly associated with parameters of systemic inflammation. In addition, they were independently associ-ated with increased risk of allograft failure long-term after kidney transplantation. Serum 3-hydroxykynurenine and 3-hydroxykynurenine-to-kynurenine ratio were also independently associated with mortality. There were no independent associations of urinary parameters of the tryptophan-kynurenine pathway with allograft failure or mortality.

(14)

7

The tryptophan-kynurenine pathway was initially thought to play an important role in preventing allograft rejection after transplantation.89,90 This was based on findings

in experimental transplantation models in small animals, where the tryptophan-ky-nurenine pathway activation was shown to induce immune tolerance.91-93 Surprisingly,

however, our study and other observational studies in human kidney transplanta-tion consistently demonstrated that tryptophan-kynurenine pathway activatransplanta-tion is associated with reduced allograft survival. For example, IDO activity, as measured by kynurenine-to-tryptophan ratio, was increased right before and during acute kidney allograft rejection.94,95 In addition, increased pre-transplantation serum kynurenine

concentration was associated with the occurrence of acute allograft rejection after kidney transplantation.96 Moreover, increased serum and urinary kynurenine early

after kidney transplantation was associated with increased serum creatinine and albuminuria two years post-transplantation.97 Finally, activation of the

tryptophan-ky-nurenine pathway has been associated with increased acute rejection rates in heart transplantation98 and increased prevalence and severity of graft-versus-host disease

after bone marrow transplantation.99

We found that serum kynurenine and 3-hydroxykynurenine were strongly associated with parameters of systemic inflammation. Indeed, the tryptophan-kynurenine path-way has been implicated in many disease states in which systemic inflammation is present.100,101 Via its role in systemic inflammation, it has also been implicated in the

development of atherosclerosis and cardiovascular disease,102,103 especially in patients

with impaired kidney function.104-106 In these patients, tryptophan-kynurenine pathway

metabolites are known to accumulate.107-112 It has been hypothesized that the increase

in serum kynurenine metabolites in patients with impaired kidney function is caused not only by reduced renal clearance of the metabolites themselves,113 but also by

increased tryptophan break-down to kynurenine by the pro-inflammatory uremic envi-ronment.109,111 Therefore, interfering in the kynurenine pathway by blocking different

steps of the pathway is currently being evaluated as potential therapeutic strategy. Initially, inhibitors of IDO gained most interest, because this enzyme catalyzes the first and rate-limiting step in the pathway.114,115 Importantly, it recently became apparent

that the IDO product kynurenine is not only metabolized to cytotoxic 3-hydroxyky-nurenine, but also to (neuro)protective kynurenic acid.114,115 Therefore, inhibitors of

the down-stream enzyme KMO were developed with the aim of reducing production of 3-hydroxykynurenine and shifting the pathway towards production of kynurenic acid.114-116

(15)

The question remains whether blocking of IDO and/or KMO could be a useful thera-peutic strategy in RTR. For this, we have to dive a bit deeper into kynurenine pathway physiology. The kynurenine pathway was originally found to play an important role in the immune response against intracellular viruses and parasites.117,118 By depleting

the essential amino acid tryptophan, which is required for growth, it protected the host against these pathogens. However, IDO was also found to suppress the immune response in experimental models.100,101 Two distinct mechanisms have been proposed

to explain the immunosuppressive function of IDO. In the first mechanism – called the “tryptophan starvation or depletion” theory, IDO activity directly reduces local tryp-tophan concentrations, thereby impeding normal proliferation in a variety of cells types and increasing their susceptibility to apoptosis. T lymphocytes in particular have been shown to be susceptible to tryptophan starvation.119,120 In the second

mecha-nism – called the “tryptophan utilization” theory, IDO activation indirectly produces immunosuppressive effects via increased formation of downstream metabolites, such as kynurenine and 3-hydroxykynurenine. These metabolites have been implicated in reduced proliferation and increased apoptosis of both T helper cells and natural killer cells.121-125 Additionally, the combined effects of tryptophan depletion and metabolite

interaction with CD8+ effector T lymphocytes reduce the capabilities of these immune response cells.124

Taken together, these proposed mechanisms suggest that activation of the kynurenine pathway could also have immunosuppressive effects in vivo. Therefore, it came as a surprise that every single clinical study in kidney and other transplant recipients showed that kynurenine pathway activation was associated with reduced allograft survival. To date, no solid explanation for this controversy has been found. One of the possible explanations could be that, in contrast to the ‘closed’ setting of experimental cell models, the human body is an open system, so tryptophan can easily be replen-ished and depletion will never take place. Moreover, in the case of inflammation, it is likely that continuously dying cells release their intracellular stocks, thereby providing additional supplies of tryptophan. In addition, in order to provide adequate immuno-suppression, tryptophan concentrations would have to fall below 1 µmol/L, which is about one fiftieth of normal tryptophan concentrations in plasma.126 The other way

around, the toxic effects of kynurenine metabolites are only apparent at concentra-tions that are 5 to 10 times higher than the expected physiological concentration.127,128

Although kynurenine and 3-hydroxykynurenine were increased in RTR with more adverse outcome in our study, their median concentrations were only 2 times higher than in RTR with more favorable outcome. Another possible explanation would be that the potentially suppressive function of kynurenine metabolites is outcompeted by the

(16)

7

overwhelming immune response during allograft rejection.89 However, although this

maybe could be true for the event of acute rejection, this would be less likely in the case of chronic systemic inflammation, which is more smoldering in nature. The third possible explanation would be that activation of the kynurenine pathway by IDO is merely a marker of ongoing systemic inflammation or immune activation. Numerous pro-inflammatory cytokines, including interferons (IFN-α, IFN-β, and IFN-γ), interleu-kins (IL-1 and IL-2), and tumor necrosis factor (TNF-α) induce the expression of IDO and also KMO.101 Therefore, measuring parameters of the kynurenine pathway may

only be useful in assessing chronic inflammation, but interfering in it would not have the expected effects. To conclude, it remains to be elucidated whether interference in or, conversely, active induction of the kynurenine pathway would be beneficial in the clinical setting of kidney transplantation, and whether we are dealing with a friend or foe, or maybe even an innocent bystander.

Towards personalized treatment in renal transplant recipients

With this thesis, we tried to make first steps towards personalized treatment of pred-nisolone-treated RTR. Although the concept of personalized medicine has received great interest over the past years, the idea dates from ancient times. Already in 400 BC, the father of early medicine, Hippocrates, said: “it is far more important to know what person the disease has, than what disease the person has”.129 Thus, the concept

that individuals with the same disease and a similar clinical presentation may have very different outcomes and need very different therapies is not novel. In modern medicine, personalized – or precision or stratified – medicine encompasses a medical model that proposes the customization of health, with medical decisions, practices and/or products being tailored to the individual patient, based on their predicted response or risk of disease.130 This is opposed to the ‘one-size-fits-all’ approach, which applies

decisions from average treatment effects observed in clinical trials to all patients. Pre-diction modeling, using information on patients’ genetic and molecular make-up, can be applied to predict different outcomes, such as the duration of survival, potential complications, response to therapy, and other outcomes. Individual genetic and molec-ular make-up would be assessed by personal ‘omics’ profiling, which uses molecmolec-ular diagnostic tools to combine information on, for example, the (epi)genome, transcrip-tome, proteome, metabolome, lipidome, and microbiome.130,131

In the field of kidney transplantation, personalized medicine, by means of assessing individual genetic and molecular make-up, could have great potential in, for exam-ple, selecting optimal donor-recipient pairs and individualizing immunosuppressive therapy, but also in predicting the probability of more complex outcomes, such as the risk of acute or chronic rejection, infections, and malignancies, which may help to

(17)

individualize immunosuppressive treatment even further.131,132 Therefore,

personal-ized immunosuppressive treatment, based on molecular profiling of each individual transplant recipient, holds great promise for optimizing the balance between thera-peutic efficacy and toxicity of treatment with these drugs, thereby potentially reducing unwanted side effects and concomitant cardiovascular risk.131 However, many

chal-lenges have to be met, such as problems with standardization, means of analyzing and interpretation of the enormous amounts of data which will come available, before the use of these complex molecular techniques can really be implemented in clinical kidney transplant care.

In the meantime, we should strive for personalized cardiovascular risk management in RTR, which encompasses the development and application of adequate transplan-tation-specific guidelines, using knowledge on the pathophysiology and risk factors specific to this patient group, rather than extrapolating guidelines from the general population or other high risk populations, in order to reduce the cardiovascular burden after kidney transplantation. In addition, as we learned from studying lifestyle inter-ventions in RTR, not only a patient’s molecular or genetic make-up determines the potential success or failure of treatment, but also his or her social environment, and beliefs and attitudes towards this treatment. Therefore, to truly personalize treatment in RTR in the future, we need an integrative systems approach which addresses both the needs and specific characteristics of this patient population, at both individual and group level. With this thesis, we show that combining lifestyle interventions with individual monitoring of immunosuppressive (i.e. corticosteroid) treatment may serve as an important first step towards personalized cardiovascular risk management in prednisolone-treated RTR.

(18)

7

REFERENCES

1. Dobrowolski LC, Bemelman FJ, van Donselaar-van der Pant KA, Hoitsma AJ, ten Berge IJ, Krediet CT: Treatment efficacy of hypertension in kidney transplant recipients in the netherlands. Neth J Med 72(5):258-263, 2014

2. Cross NB, Webster AC, Masson P, O’Connell PJ, Craig JC: Antihypertensive treatment for kidney transplant recipients. Cochrane Database Syst Rev (3):CD003598. doi(3):CD003598, 2009 3. Ojo AO: Cardiovascular complications after renal transplantation and their prevention.

Transplan-tation 82(5):603-611, 2006

4. Herzog CA, Asinger RW, Berger AK, et al: Cardiovascular disease in chronic kidney disease. A clinical update from kidney disease: Improving global outcomes (KDIGO). Kidney Int 80(6):572-586, 2011 5. Navis G, de Jong PE, Donker AJ, van der Hem GK, de Zeeuw D: Moderate sodium restriction in

hypertensive subjects: Renal effects of ACE-inhibition. Kidney Int 31(3):815-819, 1987

6. Navis G, de Jong P, Donker AJ, van der Hem GK, de Zeeuw D: Diuretic effects of angiotensin-con-verting enzyme inhibition: Comparison of low and liberal sodium diet in hypertensive patients. J Cardiovasc Pharmacol 9(6):743-748, 1987

7. Vogt L, Waanders F, Boomsma F, de Zeeuw D, Navis G: Effects of dietary sodium and hydrochloro-thiazide on the antiproteinuric efficacy of losartan. J Am Soc Nephrol 19(5):999-1007, 2008 8. Slagman MC, Waanders F, Hemmelder MH, et al: Moderate dietary sodium restriction added to

angiotensin converting enzyme inhibition compared with dual blockade in lowering proteinuria and blood pressure: Randomised controlled trial. BMJ 343:d4366, 2011

9. Kwakernaak AJ, Krikken JA, Binnenmars SH, et al: Effects of sodium restriction and hydrochlorothia-zide on RAAS blockade efficacy in diabetic nephropathy: A randomised clinical trial. Lancet Diabetes Endocrinol 2(5):385-395, 2014

10. Keyzer CA, van Breda GF, Vervloet MG, et al: Effects of vitamin D receptor activation and die-tary sodium restriction on residual albuminuria in CKD: The ViRTUE-CKD trial. J Am Soc Nephrol 28(4):1296-1305, 2017

11. McMahon EJ, Bauer JD, Hawley CM, et al: A randomized trial of dietary sodium restriction in CKD. J Am Soc Nephrol 24(12):2096-2103, 2013

12. Vegter S, Perna A, Postma MJ, Navis G, Remuzzi G, Ruggenenti P: Sodium intake, ACE inhibition, and progression to ESRD. J Am Soc Nephrol 23(1):165-173, 2012

13. Lambers Heerspink HJ, Holtkamp FA, Parving HH, et al: Moderation of dietary sodium potentia-tes the renal and cardiovascular protective effects of angiotensin receptor blockers. Kidney Int 82(3):330-337, 2012

14. Buter H, Hemmelder MH, Navis G, de Jong PE, de Zeeuw D: The blunting of the antiproteinuric efficacy of ACE inhibition by high sodium intake can be restored by hydrochlorothiazide. Nephrol Dial Transplant 13(7):1682-1685, 1998

15. Monfa E, Rodrigo E, Belmar L, et al: A high sodium intake reduces antiproteinuric response to renin-angiotensin-aldosterone system blockade in kidney transplant recipients. Nefrologia 36(5):545-551, 2016

16. van den Berg E, Geleijnse JM, Brink EJ, et al: Sodium intake and blood pressure in renal transplant recipients. Nephrol Dial Transplant 27(8):3352-3359, 2012

17. Rocchini AP: Obesity hypertension. Am J Hypertens 15(2 Pt 2):50S-52S, 2002

18. Rocchini AP: Obesity hypertension, salt sensitivity and insulin resistance. Nutr Metab Cardiovasc Dis 10(5):287-294, 2000

(19)

19. Koomans HA, Roos JC, Boer P, Geyskes GG, Mees EJ: Salt sensitivity of blood pressure in chronic renal failure. evidence for renal control of body fluid distribution in man. Hypertension 4(2):190-197, 1982

20. Curtis JJ, Luke RG, Jones P, Diethelm AG: Hypertension in cyclosporine-treated renal transplant recipients is sodium dependent. Am J Med 85(2):134-138, 1988

21. Diederich S, Scholz T, Eigendorff E, et al: Pharmacodynamics and pharmacokinetics of synthetic mineralocorticoids and glucocorticoids: Receptor transactivation and prereceptor metabolism by 11beta-hydroxysteroid-dehydrogenases. Hormone and Metabolic Research 36(6):423-429, 2004 22. Quinkler M, Jussli J, Bähr V, Pfeiffer AFH, Lepenies J, Diederich S: Effect of immunosuppressive and

other drugs on the cortisol-cortisone shuttle in human kidney and liver. Hormone and Metabolic Research = Hormon- Und Stoffwechselforschung = Hormones Et Metabolisme 39(8):555-9, 2007 23. Hoorn EJ, Walsh SB, McCormick JA, et al: The calcineurin inhibitor tacrolimus activates the renal

sodium chloride cotransporter to cause hypertension. Nat Med 17(10):1304-1309, 2011 24. Melnikov S, Mayan H, Uchida S, Holtzman EJ, Farfel Z: Cyclosporine metabolic side effects:

Associ-ation with the WNK4 system. Eur J Clin Invest 41(10):1113-1120, 2011

25. Hiremath S, Fergusson D, Doucette S, Mulay AV, Knoll GA: Renin angiotensin system blockade in kidney transplantation: A systematic review of the evidence. Am J Transplant 7(10):2350-2360, 2007 26. Apperloo AJ, de Zeeuw D, de Jong PE: A short-term antihypertensive treatment-induced fall in

glomerular filtration rate predicts long-term stability of renal function. Kidney Int 51(3):793-797, 1997

27. Holtkamp FA, de Zeeuw D, Thomas MC, et al: An acute fall in estimated glomerular filtration rate during treatment with losartan predicts a slower decrease in long-term renal function. Kidney Int 80(3):282-287, 2011

28. Cross NB, Webster AC, Masson P, O’connell PJ, Craig JC: Antihypertensives for kidney transplant recipients: Systematic review and meta-analysis of randomized controlled trials. Transplantation 88(1):7-18, 2009

29. Cross NB, Webster AC, Masson P, O’Connell PJ, Craig JC: Antihypertensive treatment for kidney transplant recipients. Cochrane Database Syst Rev (3):CD003598. doi(3):CD003598, 2009 30. Dickerson JE, Hingorani AD, Ashby MJ, Palmer CR, Brown MJ: Optimisation of antihypertensive

treatment by crossover rotation of four major classes. Lancet 353(9169):2008-2013, 1999 31. Byrd JB: Personalized medicine and treatment approaches in hypertension: Current perspectives.

Integr Blood Press Control 9:59-67, 2016

32. Sacks FM, Svetkey LP, Vollmer WM, et al: Effects on blood pressure of reduced dietary sodium and the dietary approaches to stop hypertension (DASH) diet. DASH-sodium collaborative research group. N Engl J Med 344(1):3-10, 2001

33. Kidney Disease Outcomes Quality Initiative (K/DOQI): K/DOQI clinical practice guidelines on hypertension and antihypertensive agents in chronic kidney disease. Am J Kidney Dis 43(5 Suppl 1):S1-290, 2004

34. Humalda JK, Klaassen G, de Vries H, et al: The SUBLIME approach: Efficacy and cost-effectiveness of a blended care self-management approach facilitated by E-health for dietary sodium restriction in patients with chronic kidney disease. Submitted

35. Pimenta E, Gaddam KK, Pratt-Ubunama MN, et al: Relation of dietary salt and aldosterone to urinary protein excretion in subjects with resistant hypertension. Hypertension 51(2):339-344, 2008 36. Calo LA, Puato M, Schiavo S, et al: Absence of vascular remodelling in a high angiotensin-II state

(bartter’s and gitelman’s syndromes): Implications for angiotensin II signalling pathways. Nephrol Dial Transplant 23(9):2804-2809, 2008

(20)

7

37. Nowaczynski W, Oliver WJ, Neel JV: Serum aldosterone and protein-binding variables in yanomama indians: A no-salt culture as compared to partially acculturated guaymi indians. Clin Physiol Bio-chem 3(6):289-306, 1985

38. [Anonymous]. Effectiveness of spironolactone added to an angiotensin-converting enzyme inhi-bitor and a loop diuretic for severe chronic congestive heart failure (the randomized aldactone evaluation study [RALES]). Am J Cardiol 78(8):902-907, 1996

39. Pitt B, Williams G, Remme W, et al: The EPHESUS trial: Eplerenone in patients with heart failure due to systolic dysfunction complicating acute myocardial infarction. eplerenone post-AMI heart failure efficacy and survival study. Cardiovasc Drugs Ther 15(1):79-87, 2001

40. Bianchi S, Bigazzi R, Campese VM: Long-term effects of spironolactone on proteinuria and kidney function in patients with chronic kidney disease. Kidney Int 70(12):2116-2123, 2006

41. Furumatsu Y, Nagasawa Y, Tomida K, et al: Effect of renin-angiotensin-aldosterone system triple blockade on non-diabetic renal disease: Addition of an aldosterone blocker, spironolactone, to com-bination treatment with an angiotensin-converting enzyme inhibitor and angiotensin II receptor blocker. Hypertens Res 31(1):59-67, 2008

42. Tylicki L, Rutkowski P, Renke M, et al: Triple pharmacological blockade of the renin-angiotensin-al-dosterone system in nondiabetic CKD: An open-label crossover randomized controlled trial. Am J Kidney Dis 52(3):486-493, 2008

43. Epstein M, Williams GH, Weinberger M, et al: Selective aldosterone blockade with eplerenone reduces albuminuria in patients with type 2 diabetes. Clin J Am Soc Nephrol 1(5):940-951, 2006 44. Mehdi UF, Adams-Huet B, Raskin P, Vega GL, Toto RD: Addition of angiotensin receptor blockade

or mineralocorticoid antagonism to maximal angiotensin-converting enzyme inhibition in diabetic nephropathy. J Am Soc Nephrol 20(12):2641-2650, 2009

45. Hurley HA, Haririan A: Corticosteroid withdrawal in kidney transplantation: The present status. Expert Opin Biol Ther 7(8):1137-1151, 2007

46. Srinivas TR, Meier-Kriesche HU: Minimizing immunosuppression, an alternative approach to redu-cing side effects: Objectives and interim result. Clin J Am Soc Nephrol 3 Suppl 2:S101-S116, 2008 47. Bergmann TK, Barraclough KA, Lee KJ, Staatz CE: Clinical pharmacokinetics and pharmacodynamics

of prednisolone and prednisone in solid organ transplantation. Clin Pharmacokinet 51(11):711-741, 2012

48. Boots JMM, van den Ham ,E.C.H., Christiaans MHL, van Hooff ,J.P.: Risk of adrenal insufficiency with steroid maintenance therapy in renal transplantation. Transplant Proc 34(5):1696-7, 2002 49. Broersen LHA, Pereira AM, Jorgensen JO,L., Dekkers OM: Adrenal insufficiency in corticosteroids

use: Systematic review and meta-analysis. J Clin Endocrinol Metab 100(6):2171-80, 2015 50. Bromberg JS, Alfrey EJ, Barker CF, et al: Adrenal suppression and steroid supplementation in renal

transplant recipients. Transplantation 51(2):385-390, 1991

51. Sherlock M, Behan LA, Hannon MJ, et al: The modulation of corticosteroid metabolism by hydrocor-tisone therapy in patients with hypopituitarism increases tissue glucocorticoid exposure. European Journal of Endocrinology 173(5):583-593, 2015

52. Werumeus Buning J, van Faassen M, Brummelman P, et al: Effects of hydrocortisone on the regulation of blood pressure: Results from a randomized controlled trial. The Journal of Clinical Endocrinology & Metabolism 101(10):3691-3699, 2016

53. Tomlinson JW, Stewart PM: Cortisol metabolism and the role of 11beta-hydroxysteroid dehydro-genase. Best Practice & Research.Clinical Endocrinology & Metabolism 15(1):61-78, 2001 54. Rosmalen JGM, Kema IP, Wüst S, et al: 24 H urinary free cortisol in large-scale epidemiological

(21)

47:10-6, 2014

55. Tomlinson JW, Walker EA, Bujalska IJ, et al: 11beta-hydroxysteroid dehydrogenase type 1: A tis-sue-specific regulator of glucocorticoid response. Endocr Rev 25(5):831-866, 2004

56. Cuzzola A, Mazzini F, Petri A: A comprehensive study for the validation of a LC-MS/MS method for the determination of free and total forms of urinary cortisol and its metabolites. J Pharm Biomed Anal 94:203-9, 2014

57. Remer T, Maser-Gluth C, Wudy SA: Glucocorticoid measurements in health and disease--metabolic implications and the potential of 24h urine analyses. Mini Rev Med Chem 8(2):153-170, 2008 58. Esteban NV, Loughlin T, Yergey AL, et al: Daily cortisol production rate in man determined by stable

isotope dilution/mass spectrometry. J Clin Endocrinol Metab 72(1):39-45, 1991

59. Kraan GP, Dullaart RP, Pratt JJ, Wolthers BG, Drayer NM, De Bruin R: The daily cortisol production reinvestigated in healthy men. the serum and urinary cortisol production rates are not significantly different. J Clin Endocrinol Metab 83(4):1247-1252, 1998

60. Bergrem HA, Bergrem H, Hartmann A, Hjelmesaeth J, Jenssen T: Role of prednisolone pharmacoki-netics in postchallenge glycemia after renal transplantation. Ther Drug Monit 30(5):583-590, 2008 61. Gregoor PJ, de Sevaux RG, Hene RJ, et al: Effect of cyclosporine on mycophenolic acid trough levels

in kidney transplant recipients. Transplantation 68(10):1603-1606, 1999

62. Jusko WJ, Ferron GM, Mis SM, Kahan BD, Zimmerman JJ: Pharmacokinetics of prednisolone during administration of sirolimus in patients with renal transplants. J Clin Pharmacol 36(12):1100-1106, 1996

63. Rocci ML,Jr, Tietze KJ, Lee J, Harris H, Danzeisen J, Burke JF: The effect of cyclosporine on the phar-macokinetics of prednisolone in renal transplant patients. Transplantation 45(3):656-660, 1988 64. Potter JM, McWhinney BC, Sampson L, Hickman PE: Area-under-the-curve monitoring of

predniso-lone for dose optimization in a stable renal transplant population. Ther Drug Monit 26(4):408-414, 2004

65. Bergrem H: Pharmacokinetics and protein binding of prednisolone in patients with nephrotic syn-drome and patients undergoing hemodialysis. Kidney Int 23(6):876-881, 1983

66. Bergrem H: The influence of uremia on pharmacokinetics and protein binding of prednisolone. Acta Med Scand 213(5):333-337, 1983

67. Kawai S, Ichikawa Y, Homma M: Differences in metabolic properties among cortisol, prednisolone, and dexamethasone in liver and renal diseases: Accelerated metabolism of dexamethasone in renal failure. J Clin Endocrinol Metab 60(5):848-854, 1985

68. Reece PA, Disney AP, Stafford I, Shastry JC: Prednisolone protein binding in renal transplant patients. Br J Clin Pharmacol 20(2):159-162, 1985

69. Stuck AE, Frey BM, Frey FJ: Kinetics of prednisolone and endogenous cortisol suppression in the elderly. Clin Pharmacol Ther 43(4):354-362, 1988

70. Quax RA, Manenschijn L, Koper JW, et al: Glucocorticoid sensitivity in health and disease. Nat Rev Endocrinol 9(11):670-686, 2013

71. Henley D, Lightman S, Carrell R: Cortisol and CBG - getting cortisol to the right place at the right time. Pharmacol Ther 166:128-135, 2016

72. Chapman K, Holmes M, Seckl J: 11beta-hydroxysteroid dehydrogenases: Intracellular gate-keepers of tissue glucocorticoid action. Physiol Rev 93(3):1139-1206, 2013

73. Bartholome B, Spies CM, Gaber T, et al: Membrane glucocorticoid receptors (mGCR) are expressed in normal human peripheral blood mononuclear cells and up-regulated after in vitro stimulation and in patients with rheumatoid arthritis. FASEB J 18(1):70-80, 2004

(22)

7

glucocorticoid resistance. Ann N Y Acad Sci 1069:1-9, 2006

75. Koper JW, van Rossum EF, van den Akker EL: Glucocorticoid receptor polymorphisms and haploty-pes and their expression in health and disease. Steroids 92:62-73, 2014

76. N’Gankam V, Uehlinger D, Dick B, Frey BM, Frey FJ: Increased cortisol metabolites and redu-ced activity of 11beta-hydroxysteroid dehydrogenase in patients on hemodialysis. Kidney Int 61(5):1859-1866, 2002

77. Boonen E, Bornstein SR, Van dB: New insights into the controversy of adrenal function during critical illness. The Lancet.Diabetes & Endocrinology 3(10):805-15, 2015

78. Boonen E, Vervenne H, Meersseman P, et al: Reduced cortisol metabolism during critical illness. N Engl J Med 368(16):1477-88, 2013

79. Vogt B, Dick B, N’Gankam V, Frey FJ, Frey BM: Reduced 11beta-hydroxysteroid dehydrogenase activity in patients with the nephrotic syndrome. J Clin Endocrinol Metab 84(0021-972; 2):811-814, 1999

80. Reese PP, Bloom RD, Trofe-Clark J, et al: Automated reminders and physician notification to pro-mote immunosuppression adherence among kidney transplant recipients: A randomized trial. Am J Kidney Dis 69(3):400-409, 2017

81. Zhu Y, Zhou Y, Zhang L, Zhang J, Lin J: Efficacy of interventions for adherence to the immunosup-pressive therapy in kidney transplant recipients: A meta-analysis and systematic review. J Investig Med 65(7):1049-1056, 2017

82. Bakri RS, Afzali B, Covic A, et al: Cardiovascular disease in renal allograft recipients is associated with elevated sialic acid or markers of inflammation. Clin Transplant 18(2):201-204, 2004 83. Dahle DO, Mjoen G, Oqvist B, et al: Inflammation-associated graft loss in renal transplant recipients.

Nephrol Dial Transplant 26(11):3756-3761, 2011

84. van Ree RM, Gross S, Zelle DM, et al: Influence of C-reactive protein and urinary protein excretion on prediction of graft failure and mortality by serum albumin in renal transplant recipients. Trans-plantation 89(10):1247-1254, 2010

85. van Ree RM, Oterdoom LH, de Vries AP, et al: Elevated levels of C-reactive protein independently predict accelerated deterioration of graft function in renal transplant recipients. Nephrol Dial Trans-plant 22(1):246-253, 2007

86. Takikawa O, Yoshida R, Kido R, Hayaishi O: Tryptophan degradation in mice initiated by indoleamine 2,3-dioxygenase. J Biol Chem 261(8):3648-3653, 1986

87. Ozaki Y, Edelstein MP, Duch DS: Induction of indoleamine 2,3-dioxygenase: A mechanism of the antitumor activity of interferon gamma. Proc Natl Acad Sci U S A 85(4):1242-1246, 1988

88. Yamazaki F, Kuroiwa T, Takikawa O, Kido R: Human indolylamine 2,3-dioxygenase. its tissue distri-bution, and characterization of the placental enzyme. Biochem J 230(3):635-638, 1985

89. Lob S, Konigsrainer A: Role of IDO in organ transplantation: Promises and difficulties. Int Rev Immunol 28(3-4):185-206, 2009

90. Mulley WR, Nikolic-Paterson DJ: Indoleamine 2,3-dioxygenase in transplantation. Nephrology (Carl-ton) 13(3):204-211, 2008

91. Lin YC, Chen CL, Nakano T, et al: Immunological role of indoleamine 2,3-dioxygenase in rat liver allograft rejection and tolerance. J Gastroenterol Hepatol 23(7 Pt 2):e243-50, 2008

92. Grohmann U, Orabona C, Fallarino F, et al: CTLA-4-ig regulates tryptophan catabolism in vivo. Nat Immunol 3(11):1097-1101, 2002

93. Cook CH, Bickerstaff AA, Wang JJ, et al: Spontaneous renal allograft acceptance associated with “regulatory” dendritic cells and IDO. J Immunol 180(5):3103-3112, 2008

(23)

through IDO metabolism evaluation. Kidney Int 71(1):60-67, 2007

95. Zhao X, Chen J, Ye L, Xu G: Serum metabolomics study of the acute graft rejection in human renal transplantation based on liquid chromatography-mass spectrometry. J Proteome Res 13(5):2659-2667, 2014

96. Lahdou I, Sadeghi M, Daniel V, et al: Increased pretransplantation plasma kynurenine levels do not protect from but predict acute kidney allograft rejection. Hum Immunol 71(11):1067-1072, 2010 97. Vavrincova-Yaghi D, Seelen MA, Kema IP, et al: Early posttransplant tryptophan metabolism predicts

long-term outcome of human kidney transplantation. Transplantation 2015

98. Suarez-Fuentetaja N, Domenech-Garcia N, Paniagua-Martin MJ, et al: Indoleamine, 2-3 dioxyge-nase activity could be an early marker of graft rejection in heart transplantation. Transplant Proc 44(9):2645-2648, 2012

99. Xu J, Wei J, Zhu X, et al: Increased plasma indoleamine 2,3-dioxygenase activity and interfe-ron-gamma levels correlate with the severity of acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 19(2):196-201, 2013 100. Kolodziej LR, Paleolog EM, Williams RO: Kynurenine metabolism in health and disease. Amino Acids

41(5):1173-1183, 2011

101. Wang Q, Liu D, Song P, Zou MH: Tryptophan-kynurenine pathway is dysregulated in inflammation, and immune activation. Front Biosci (Landmark Ed) 20:1116-1143, 2015

102. Pertovaara M, Raitala A, Juonala M, et al: Indoleamine 2,3-dioxygenase enzyme activity correlates with risk factors for atherosclerosis: The cardiovascular risk in young finns study. Clin Exp Immunol 148(1):106-111, 2007

103. Niinisalo P, Raitala A, Pertovaara M, et al: Indoleamine 2,3-dioxygenase activity associates with cardiovascular risk factors: The health 2000 study. Scand J Clin Lab Invest 68(8):767-770, 2008 104. Pawlak K, Brzosko S, Mysliwiec M, Pawlak D: Kynurenine, quinolinic acid--the new factors linked to

carotid atherosclerosis in patients with end-stage renal disease. Atherosclerosis 204(2):561-566, 2009

105. Pawlak K, Domaniewski T, Mysliwiec M, Pawlak D: The kynurenines are associated with oxidative stress, inflammation and the prevalence of cardiovascular disease in patients with end-stage renal disease. Atherosclerosis 204(1):309-314, 2009

106. Pawlak K, Domaniewski T, Mysliwiec M, Pawlak D: Kynurenines and oxidative status are indepen-dently associated with thrombomodulin and von willebrand factor levels in patients with end-stage renal disease. Thromb Res 124(4):452-457, 2009

107. Pawlak D, Pawlak K, Malyszko J, Mysliwiec M, Buczko W: Accumulation of toxic products degradation of kynurenine in hemodialyzed patients. Int Urol Nephrol 33(2):399-404, 2001

108. Saito K, Fujigaki S, Heyes MP, et al: Mechanism of increases in L-kynurenine and quinolinic acid in renal insufficiency. Am J Physiol Renal Physiol 279(3):F565-72, 2000

109. Schefold JC, Zeden JP, Fotopoulou C, et al: Increased indoleamine 2,3-dioxygenase (IDO) activity and elevated serum levels of tryptophan catabolites in patients with chronic kidney disease: A possible link between chronic inflammation and uraemic symptoms. Nephrol Dial Transplant 24(6):1901-1908, 2009

110. Mutsaers HA, Masereeuw R, Olinga P: Altered tryptophan metabolism and CKD-associated fatigue. Kidney Int 86(5):1061-1062, 2014

111. Toyohara T, Akiyama Y, Suzuki T, et al: Metabolomic profiling of uremic solutes in CKD patients. Hypertens Res 33(9):944-952, 2010

112. Yilmaz N, Ustundag Y, Kivrak S, et al: Serum indoleamine 2,3 dioxygenase and tryptophan and kynurenine ratio using the UPLC-MS/MS method, in patients undergoing peritoneal dialysis,

(24)

hemo-7

dialysis, and kidney transplantation. Ren Fail 38(8):1300-1309, 2016

113. Moller SE: Pharmacokinetics of tryptophan, renal handling of kynurenine and the effect of nicoti-namide on its appearance in plasma and urine following L-tryptophan loading of healthy subjects. Eur J Clin Pharmacol 21(2):137-142, 1981

114. Dounay AB, Tuttle JB, Verhoest PR: Challenges and opportunities in the discovery of new therapeu-tics targeting the kynurenine pathway. J Med Chem 58(22):8762-8782, 2015

115. Zadori D, Veres G, Szalardy L, et al: Inhibitors of the kynurenine pathway as neurotherapeutics: A patent review (2012-2015). Expert Opin Ther Pat 26(7):815-832, 2016

116. Smith JR, Jamie JF, Guillemin GJ: Kynurenine-3-monooxygenase: A review of structure, mechanism, and inhibitors. Drug Discov Today 21(2):315-324, 2016

117. Pfefferkorn ER: Interferon gamma blocks the growth of toxoplasma gondii in human fibroblasts by inducing the host cells to degrade tryptophan. Proc Natl Acad Sci U S A 81(3):908-912, 1984 118. Byrne GI, Lehmann LK, Landry GJ: Induction of tryptophan catabolism is the mechanism for

gam-ma-interferon-mediated inhibition of intracellular chlamydia psittaci replication in T24 cells. Infect Immun 53(2):347-351, 1986

119. Mellor AL, Munn DH: Tryptophan catabolism and T-cell tolerance: Immunosuppression by starva-tion? Immunol Today 20(10):469-473, 1999

120. Orabona C, Grohmann U: Indoleamine 2,3-dioxygenase and regulatory function: Tryptophan star-vation and beyond. Methods Mol Biol 677:269-280, 2011

121. Belladonna ML, Puccetti P, Orabona C, et al: Immunosuppression via tryptophan catabolism: The role of kynurenine pathway enzymes. Transplantation 84(1 Suppl):S17-20, 2007

122. Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB: Tryptophan-derived catabo-lites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J Exp Med 196(4):459-468, 2002

123. Kai S, Goto S, Tahara K, Sasaki A, Kawano K, Kitano S: Inhibition of indoleamine 2,3-dioxygenase suppresses NK cell activity and accelerates tumor growth. J Exp Ther Oncol 3(6):336-345, 2003 124. Quan J, Tan PH, MacDonald A, Friend PJ: Manipulation of indoleamine 2,3-dioxygenase (IDO) for

clinical transplantation: Promises and challenges. Expert Opin Biol Ther 8(11):1705-1719, 2008 125. Terness P, Bauer TM, Rose L, et al: Inhibition of allogeneic T cell proliferation by indoleamine

2,3-dioxygenase-expressing dendritic cells: Mediation of suppression by tryptophan metabolites. J Exp Med 196(4):447-457, 2002

126. Munn DH, Shafizadeh E, Attwood JT, Bondarev I, Pashine A, Mellor AL: Inhibition of T cell prolifera-tion by macrophage tryptophan catabolism. J Exp Med 189(9):1363-1372, 1999

127. Lee GK, Park HJ, Macleod M, Chandler P, Munn DH, Mellor AL: Tryptophan deprivation sensitizes activated T cells to apoptosis prior to cell division. Immunology 107(4):452-460, 2002

128. Munn DH, Sharma MD, Baban B, et al: GCN2 kinase in T cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-dioxygenase. Immunity 22(5):633-642, 2005 129. Hippocrates: Hippocrates, volume I. Cambridge, Harvard University Press, 1995

130. Schleidgen S, Klingler C, Bertram T, Rogowski WH, Marckmann G: What is personalized medicine: Sharpening a vague term based on a systematic literature review. BMC Med Ethics 14:55-6939-14-55, 2013

131. Naesens M, Anglicheau D: Precision transplant medicine: Biomarkers to the rescue. J Am Soc Nep-hrol 2017

132. Goldfarb-Rumyantzev AS: Personalized medicine and prediction of outcome in kidney transplant. Am J Kidney Dis 56(5):817-819, 2010

(25)

Referenties

GERELATEERDE DOCUMENTEN

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly

In this study, we assessed the exposure to the most common non-persistent EDCs including five parabens, three bisphenols and thirteen metabolites of in total eight different

Specific activities (Aspec. ) and conversions (C) for the light-driven imine reduction of substrates (S) 1 a–1 h using three different recombinant Synechocystis strains..

People who are aware of the persuasive intent of explicit celebrity endorsement, are more likely to activate their persuasion knowledge, this especially happens when people are

Financial support by the University of Groningen, University Medical Center Groningen, Groningen Graduate School of Medical Sciences, Dutch Kidney Foundation, Neder-

Therefore, in order to improve long-term outcome after kidney transplantation, we are in need of comprehensive strategies to reduce increased cardiovascular risk, ideally

The aim of this randomized cross-over clinical trial is therefore to assess the effects of dietary sodium restriction on BP and urinary albumin excretion (UAE) in stable

Patients with primary hyperaldosteronism, or presumed idiopathic hyperaldosteronism have an increased risk of stroke, myocardial infarction, and atrial fibrillation and have