• No results found

Hypertension after kidney transplantation - Thesis (complete)

N/A
N/A
Protected

Academic year: 2021

Share "Hypertension after kidney transplantation - Thesis (complete)"

Copied!
151
0
0

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

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

Hypertension after kidney transplantation

Dobrowolski, L.C.

Publication date

2016

Document Version

Final published version

Link to publication

Citation for published version (APA):

Dobrowolski, L. C. (2016). Hypertension after kidney transplantation.

General rights

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 personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

Hypertension after Kidney Transplantation

H

yp

ert

ens

ion

a

fte

r K

id

ne

y T

rans

pla

nta

tion

Linn

D

ob

ro

w

ols

(3)
(4)

Linn Charlotte Dobrowolski

HYPERTENSION AFTER

KIDNEY TRANSPLANTATION

(5)

PhD thesis, University of Amsterdam, The Netherlands ISBN: 978–94–6169–975-6

Online: http://dare.uva.nl

© 2016 Linn Charlotte Dobrowolski, Amsterdam, The Netherlands

All rights reserved. No part of this thesis may be reproduced, stored or transmitted in any form or by any means without prior permission of the author.

Cover design: Magnolia Branch with Four Flowers, anonymous, 1910-1925 (X-ray, gelatin silver print) Rijksmuseum Amsterdam

Lay-out: Myrthe Yeh, Linn Dobrowolski Paranymphs: Sofie Jansen, Sophie Jutte

Printed by: Optima Grafische Communicatie B.V. Rotterdam

The studies presented in this thesis have been prepared and conducted at the Department of Internal Medicine, division of Nephrology and Kidney Transplantation, Academic Medical Center at the University of Amsterdam, The Netherlands and partly at the Department of Internal Medicine, division of Nephrology and Kidney Transplantation at the University of Groningen, The Netherlands.

The Dutch Kidney Foundation (Innovation Grant IP-11.40) financially supported the research published in this thesis, which is gratefully acknowledged.

Financial support by the Dutch Heart Foundation for the publication of this thesis is gratefully acknowledged.

Financial support for the printing of this thesis was gratefully received from the Academic Medical Center Amsterdam.

Further financial support for the printing of this thesis was kindly provided by Astellas Pharma B.V, Chiesi Pharmaceuticals B.V., Chipsoft B.V. and Shire Pharmaceuticals Plc.

(6)

ACADEMISCH PROEFSCHRIFT ter verkrijging van de graad van doctor

aan de Universiteit van Amsterdam op gezag van de Rector Magnificus

prof. dr. ir. K.I.J. Maex

ten overstaan van een door het College voor Promoties ingestelde commissie, in het openbaar te verdedigen in de Aula der Universiteit

op woensdag 7 december 2016, te 11.00 uur

door

Linn Charlotte Dobrowolski geboren te Rotterdam

(7)

Promotores: Prof. dr. R.J.M. ten Berge Universiteit van Amsterdam Prof. dr. F.J. Bemelman Universiteit van Amsterdam Co-promotor: Dr. C.T.P. Krediet Universiteit van Amsterdam Overige leden: Prof. dr. T. van Gelder Erasmus Universiteit Rotterdam

Prof. dr. J. Booij Universiteit van Amsterdam Prof. dr. G. Navis Rijksuniversiteit Groningen Prof. dr. J.J. Homan van der Heide Universiteit van Amsterdam Prof. dr. E.S.G. Stroes Universiteit van Amsterdam Dr. P.J. Blankestijn Universiteit Utrecht

(8)

You are bound to be successful, bound to be successful. S.N. Goenka

(9)

123I-mIBG 123I-meta-Iodobenzylguanidine

ABPM ambulatory blood pressure measurement ACEi angiotensin converting enzyme inhibitor ARB angiotensin II receptor blocker

BP blood pressure CKD chronic kidney disease CNI calcineurin inhibitor

CTS Collaborative Transplant Study CVD cardiovascular disease

eGFR estimated glomerular filtration rate ESRD end stage renal disease

HTN hypertension IQR interquartile range

KDIGO kidney disease improving global outcomes KTR kidney transplant recipient

MDRD modification of diet in renal disease MSNA muscle sympathetic nerve activity NE norepinephrine

NOTR Netherlands Organ Transplantation Registry ODT once day prolonged release tacrolimus p.i. post-injection

PRA plasma renin activity

RAAS renin-angiotensin-aldosterone system

RDN catheter-based renal sympathetic denervation RHT resistant hypertension

RICH-Q Renal Insufficiency Therapy in Children: Quality Assessment and Improvement ROI region of interest

SNA sympathetic nerve activity

SPECT single photon emission computed tomography TDT twice day tacrolimus

(10)

Chapter 1 General introduction and outline of the thesis 9 Chapter 2 Treatment efficacy of hypertension in kidney transplant

recipients in The Netherlands

29

Chapter 3 Epidemiology and management of hypertension in paediatric and young adult kidney transplant recipients in The Netherlands

41

Chapter 4 Effects of dietary sodium restriction in kidney transplant recipients treated with renin-angiotensin-aldosterone system blockade: a randomized clinical trial

59

Chapter 5 Blood pressure dipping in kidney transplant recipients on regular and prolonged release tacrolimus

77

Chapter 6 Kidney transplant ¹²³I-mIBG scintigraphy and functional sympathetic reinnervation

89

Chapter 7 Renal sympathetic nerve activity after catheter-based renal denervation.

103

Chapter 8 Renal denervation for drug-resistant hypertension after kidney transplantation

119

Chapter 9 General discussion and perspectives 128

Summary of the thesis 131

Samenvatting (Dutch summary) 134

Appendices Supplementary documents 140

Portfolio 142

Bibliography 144

Curriculum Vitae 145

(11)
(12)

Linn C. Dobrowolski, Frederike J. Bemelman,

Ineke J. ten Berge, C.T. Paul Krediet

CHAPTER 1

GENERAL INTRODUCTION AND

OUTLINE OF THE THESIS

(13)

GENERAL INTRODUCTION Kidney transplantation

Epidemiology and survival rates

Since the first kidney transplantation in 1950 and the introduction of robust immunosuppressive regimens in the 1970s, kidney transplantation has evolved as the most durable and quality of life improving renal replacement modality. Five-year survival probability for a kidney transplant recipient (KTR) is 86.8%, compared to 53% for a patient on dialysis (Figure 1a).1 The expected unadjusted remaining lifetime for

a 40 year old patient on dialysis is 11.5 years, compared to 25.5 years of a transplant recipient (Figure 1b).1

Successful transplantation results from various partially modifiable factors, including surgical techniques, donor characteristics and effective immunosuppression.2

Post-transplant complications involve predominantly manifestation of cardiovascular disease (CVD), allograft rejection, infections attributable to compromised immune status and development of malignancies.

Most kidney transplant recipients die with a functioning allograft, i.e. without returning to dialysis and the main cause of death of KTRs is of cardiovascular disease.3-6 But

non-fatal cardiovascular disease is also highly prevalent in kidney transplant recipients (Figure 1c). Both fatal and non-fatal cardiovascular disease such as ischemic heart disease and stroke in KTRs are intertwined with transplant related factors such as allograft and donor characteristics and comorbid conditions.

Cardiovascular disease and hypertension after transplantation

Adult KTRs have an annual 50-fold increased risk for a cardiovascular event compared to the general population.7 In pediatric KTRs the risk of dying from a CVD event is also

40 times higher than in their healthy peers.8 CVD accounts for approximately 36% of

deaths in kidney transplant recipients.

Hypertension and graft loss and mortality

There is convincing (i.e. grade Ia) evidence that blood pressure (BP) reduction substantially decreases the risk for CVD events and death including coronary events, stroke, heart failure and end stage renal disease (ESRD) in the general population.9-13 For KTRs no

such level of evidence is available but observational data suggest that lowering blood pressure in hypertensive KTRs improves graft survival and lowers patients mortality.14

Landmark data from Opelz and colleagues show a strong association between BP and both graft and recipient survival (Figure 1d).15 Any 10 mmHg increase in systolic

BP is associated with a risk of graft failure up to 15%.16 Also, patient survival rates

are superior in patients without hypertension: each 10 mmHg rise in systolic blood pressure > 140 mmHg, is associated with a hazard ratio of 1.18 (95% CI 1.12 to 1.23) even after adjustment for allograft function.15-17

(14)

These data also suggest that the damage to the allograft caused by prolonged elevated BP is partially reversible. Graft survival improved in patients with high systolic blood pressure (SBP) at 1 and 3 years and a normalised SBP (< 140 mmHg) at year 5, compared to patients who remained hypertensive in the 1st, 3rd and in the 5th year after

transplantation. Lowering BP can provide significant benefits for graft and patient survival even several years after transplantation, which stresses the importance of BP control.18

Randomized controlled intervention studies in kidney transplant recipients that assess the effect of BP control, various BP targets or different antihypertensive regimens are scarce. Therefore, extrapolation of data from randomised trials on the effects of hypertension on CVD in chronic kidney disease (CKD), combined with data for large observational studies (KTR database cohorts) provides the rational for strict blood pressure control in the transplant population.15,18

Figure 1. Figure 1a: Survival by modality. European cohort 2003-2007 ERA EDTA Rep 2012).

All = transplantation and dialysis patients, Figure 1b: Life expectancy, Figure 1c: Mortality rate by cause of death, Figure 1d: Graft survival (data from Opelz et al. Kidney Int 1998).1,6,15

(15)

Definitions of hypertension and blood pressure targets after kidney transplantation

Definition of hypertension and blood pressure targets

For the general population < 60 years, adults with diabetes as well as adults with non-diabetic chronic kidney disease, hypertension is defined as systolic BP ≥ 140 mmHg and/or diastolic BP ≥ 90 mmHg. Blood pressure goals in adults > 60 years are < 150/90 mmHg.9,19 For children < 18 years, BP ≥ 95th percentile for gender, age and height

is regarded as hypertension. For kidney transplant recipients, BP target is defined at < 130/80 mmHg by the guidelines of the Kidney Disease: Improving Global Outcomes (KDIGO) because they are classified in the high-risk for CVD. For children, guidelines vary between BP thresholds of p90 and p95.14,20,21

‘Resistant hypertension’ is the term to indicate failure to reach BP targets despite the use at minimum three different antihypertensive agents, including a diuretic.22 Although

many patients seem to meet this condition, by far the most patients have secondary hypertension or are incompliant to medication or lifestyle changes.22-24 ‘Apparent

resistant hypertension’ is therefore the preferred wording. Also, rare causes of secondary hypertension should not be overlooked: Figure 2 shows a rare but treatable cause of treatment resistant hypertension in a KTR.25

Diagnosis

Diagnosing hypertension using home BP measurement or ambulatory BP monitoring (ABPM) is preferred since reliance exclusively on office-based BP measurements would result in under- or overestimation of KTRs with hypertension.26-28 BP self-measurements

can improve adherence. ABPM offers 24 hour registrations and is required when ‘white coat’ hypertension, drug resistance, symptomatic hypotension during antihypertensive therapy, or autonomic dysfunction is suspected.9

Nocturnal blood pressure

In the treatment of hypertension, nocturnal blood pressure is also of importance. Blood pressure falls with > 10% during night-time. However, as a result of altered circadian regulation, the absence of a nocturnal drop, so-called 'non-dipping', is common amongst ESRD and renal transplant patients.27,29,30 A non-dipping pattern

is an additional, independent risk factor for CVD and all-cause mortality in essential hypertensives, ESRD patients and KTRs.27,31-34 In the long term, successful transplantation

can significantly improve circadian BP profiles.35

Prevalence of post-transplant hypertension

Due to the heterogeneity in literature of definitions for hypertension in kidney transplant recipients, its true prevalence can only be estimated. Nevertheless, hypertension is observed in the majority of kidney transplant recipients and BP control rates are low. Overall prevalence in adults reaches up to 85%.16,36,37 Only 8% of patients treated with

(16)

are derived from research dated more than 20 years ago.15 Of the paediatric transplant

recipients, approximately 60-70% meet hypertension criteria.21,38-40

Pathophysiology of hypertension after kidney transplantation

Factors contributing to the pathophysiology of hypertension as well as traditional factors for hypertension are listed in Table 1. During the progression of CKD, nephrogenic factors, atherosclerosis and/or arteriosclerosis cause the increase in arterial stiffness and contribute to hypertension.41,42 The increased arterial stiffness is largely irreversible

and although transplantation prevents progression, the KTR maintains this vascular status.43,44 The allograft factors and effects of immunosuppressive agents are additional

contributing hypertension inducing factors in KTRs. Hypertension occurring in the peri-operative period is often caused by volume overload, delayed graft function and higher immunosuppressive dosages.

Figure 2. An unusual case of therapy resistant hypertension. Therapy resistant hypertension

resulting from hypoperfusion of the kidney allograft: an abdominal aortic stenosis located at the level of the superior mesenteric artery caused a severe stenotic segment (arrowhead) of the infrarenal aorta that induced hypoperfusion of the lower body including the kidney allograft (located in the right ileac fossa). Collateral circulation from the proximal to the distal aorta had developed via the superior mesenteric artery, flowing retrogradely through the hypertrophic inferior mesenteric artery (arrow).25

(17)

Donor factors

A recipient who has been transplanted with the kidney of a hypertensive donor is more likely to develop hypertension after transplantation, suggesting a genetic predisposition in the donor kidney.45-48 This theory of the primacy of the kidney is well shown by

studies that observed remission of hypertension in patients after transplantation with a normotensive donor.46

Immunosuppressive agents

After transplantation hypertension is attenuated by immunosuppressive regimens (Table 1).49,50 The corner stones of modern immunosuppressive regimens are

corticosteroids and the calcineurin inhibitors (CNI) (i.e. cyclosporine and tacrolimus). Corticosteroids cause hypertension only by both increased renal sodium absorption and alterations in the vascular tone causing increased systemic vascular resistance.51-53

Although CNIs are strong pharmacological agents preventing allograft rejection, paradoxically, exposure is associated with nephrotoxicity, which causes irreversible, progressive tubulo-interstitial fibrosis and glomerulosclerosis possibly via blood pressure and intra-renal hemodynamic effects.54 CNIs cause sodium retention and also reduce

vasodilator nitric oxide and consequently alterations in vascular diameter and flow occur.55 These effects may promote the progression of hypertension. Besides the sodium

dependency, effects on vascular tone and elevation of sympathetic nerve activity are involved in the pathogenesis of CNI induced hypertension.56-58 The more frequently

prescribed tacrolimus has a more favourable side effect pattern than cyclosporine.59,60

Sympathetic nerve activity

The renal sympathetic nervous system is thought to play an important role in blood pressure regulation. The tubules, juxtaglomerular apparatus and the vasculature are innervated by sympathetic nerves arising from the spinal cord in the ganglia T11 to L1.61

Postganglionic nerves cross the celiac and aorto-renal ganglia and reach the kidney through the walls of the extra- and intrarenal arteries. Elevated sympathetic nerve activity (SNA) induces changes in tubular sodium and water reabsorption, mediates renin-angiotensin-aldosterone system (RAAS) activation by the juxtaglomerular apparatus and adjusts glomerular filtration by vasoconstriction (Figure 3). Kidney failure is associated with a rise in SNA, manifested in early stages of CKD and is directly related to disease severity.62,63 Kidney transplantation itself has no deactivating effect on SNA. However,

bilateral nephrectomy does decrease SNA.64,65 This suggests that a neural factor arising

from the native kidneys is a centrally acting, hypertension inducing element. Kidney allografts are surgically fully denervated at the time of transplantation, but post-transplant nerve restoration is histological confirmed in both experimental and human studies.66,67

Reinnervation in time after transplantation is a continuous process, but whether nerve restoration after transplantation has beneficial or detrimental effects on graft function is yet unknown. Since overactive sympathetic drive may contribute to hypertension, (re)innervation of the kidney allograft could potentially contribute to progression of hypertension. Remarkably, renal sympathetic innervation in CKD has recently been

(18)

proposed as a supplementary factor causing renal fibrosis.68 The immunomodulatory

capacities of SNA and the possible driving factor in low-grade inflammation open a new window on nephropathy and are subject of current research. A potential link between sympathetic driven neurotransmission and fibrotic, inflammatory processes was determined in mouse models with chronic kidney injury: innervated kidneys with CKD expressed more fibrosis than chemically denervated kidneys.68 These experimental

data give ground to the idea that that renal innervation could possibly contribute to progression of allograft nephropathy.

Therapy for post-transplant hypertension

To reach BP targets in kidney transplant recipients, a uniform management strategy is essential that consists of modifying lifestyle, adequate antihypertensive medication and therapy adherence.69 Previously, bilateral nephrectomy of the native kidneys was a last

resort therapy for refractory hypertension in ESRD patients.70,71 However, over the last

Table 1. Factors contributing to post-transplant hypertension

Transplantation related Transplantation unrelated

Severe ischemia-reperfusion injury Native kidney disease Warm and Cold ischemia Systemic sympathetic activity

Delayed graft function RAAS activation (by native kidneys, sodium intake, SNA) Volume status: hypervolemia Pre-existing hypertension & left ventricular hypertrophy

Primary hyperaldosteronism

Donor: Vascular abnormalities causing renal hypoperfusion

Sex (female > male)

Donor age Traditional risk factors for hypertension:

Donor hypertension Genetic predisposition to hypertension Size of the kidney allograft Sex (male > female)

Genetic polymorphisms Age

Obesity**

Impaired graft function mostly due to: Hypercholesterolaemia

Chronic allograft nephropathy Obstructive sleep apnoea syndrome (OSAS) Thrombotic microangiopathy High sodium – diet

Rejection* Smoking

Recurrent / de novo kidney disease in graft Alcohol intake

Graft artery stenosis Chronic psychological stress Arteriovenous fistula after biopsy

Obstructive uropathy with hydronephrosis Immunosuppressive regimen:

Glucocorticosteroids Calcineurin-inhibitors / toxicity (cyclosporine > tacrolimus)

* acute, chronic, cellular or antibody mediated; ** may be transplant-related since BMI increases after transplantation

(19)

decades the procedure has been barely performed due to the risks and development of more potent antihypertensive drugs.16

Pharmacological approaches are the current foundations of antihypertensive therapy. Dose modification or alteration of immunosuppressive drugs should also be considered at all times. Steroid avoidance reduces the need for antihypertensive drugs, without affecting risk of graft loss.72,73 Tacrolimus has less hypertension inducing effects than

cyclosporine and conversion from cyclosporine to tacrolimus significant decreases BP.74-77

CNI-free regimens may be superior for amelioration of cardiovascular risk profiles, but currently they are only restricted for specific patient groups.78,79

Lifestyle

Several life style interventions have been proven to be effective in the treatment of hypertension. These include: weight reduction (to ideal body weight), physical activity (30 min per day 5 times a week), cessation of smoking, sodium restriction (to 90 mmol/

Figure 3. Renal sympathetic nerve activity and blood pressure regulation. Afferent and efferent

neural factors arising from diseased native kidneys contribute to hypertension via RAAS activation and increased vascular resistance. Abbreviations: SNA = sympathetic nerve activity; Ang II = angiotensin II; BP = blood pressure.

(20)

day) and moderation of the use of alcohol (maximum of 2 alcohol containing drinks a day).80,81 Obesity adversely affects graft outcome. Weight loss is of special interest to

KTRs because increase in body mass composition, especially body-fat, is a common finding after transplantation.82,83 Causal factors of weight gain in KTRs include physical

and metabolic effects of restored kidney function and effects of corticosteroids on food intake.84 Low physical activity in KTRs is strongly associated with a higher CVD risk

and all-cause mortality, stressing the necessity for physical activity.85 Sodium restriction

needs to be an integrated part of each antihypertensive therapy. The KDIGO guideline on this aspect prescribes a target daily sodium excretion of < 90 mmol. However, 85% of Dutch kidney transplant recipients exceed this target with a daily urinary sodium excretion of 150-176 mmol/24 h.86-88

Pharmacology

The choice of antihypertensive pharmacotherapy is based on co-morbidity, efficacy, tolerability and interactions with other drugs. Predominantly, the calcium channel blockers (CCB), beta- and alpha-1 blockers, angiotensin converting enzyme inhibitors (ACEi) and angiotensin receptor blocker (ARB) are prescribed. CCBs are often first line therapy because they counteract vasoconstrictive effects of CNIs.89 Diuretics are

beneficial since CNI induced hypertension is sodium dependent, however they are cautiously used.90,91 Although the use of ARBs and ACEi are controversial due to

intrinsic effects of glomerular filtration rate that may mimic rejection, their use results in clinically significant reduction in proteinuria.92 The effects on graft or patient survival

are unknown.93

Adherence to medication

Low medication compliance, observed in of 30-50% of the KTRs, is a modifiable behavioural factor that negatively influences graft and patient survival.94-96 In adult KTRs,

adherence to immunosuppressive agents is better than to non-immunosuppressive agents.97 Puberty as well as the transition from the small-scale, child-and-parent focused

paediatric care at the age of 18, to a large-scale, adult care program can be accompanied with significant effects of non-compliance to medication regimens.98-100 Transition occurs

in a period in which adolescents grow into young adults, increase their independency from parents and can experience less monitoring of their medical routine.101

Adherence can be improved using fixed-combined agents and by limiting the frequency of medication-intake.102 Non-adherence rates falls from 79% with taking medications

once daily to 51% with 4 times daily dosing.103 A cohesive transition program in which

paediatric and adult healthcare services are integrated may improve compliance.104

Encouraging prescription of combination/fixed-dose antihypertensive agents may increase compliance and blood pressure regulation.101,105

Innovative hypertension therapies

Because of the unmet needs for controlling BP as discussed above, over recent years innovative interventional strategies have been developed, while antihypertensive

(21)

pharmacological developments decline.106 As an advantage, interventional therapies

omit medication adherence pitfalls. Furthermore, regulation for medical devices development has been more lenient than those for pharmacological agents, therefore the devices are easier launched.

Figure 4 provides an overview of recently developed antihypertensive intervention therapies. Electric stimulation of the baro-receptors, ‘baro-receptor activation therapy’, using an implantable device may lower BP (Figure 4a).107-109 Furthermore, in a selected

group of patients, short term effects on BP reduction have been achieved using an mechanical implant in the carotid sinus that can enhance the baroreflex response and amplifies the blood pressure lowering response (Figure 4b).110-112 Also surgical

‘neurovascular decompression’ of looping arteries that compress rostral ventrolateral medullae, have shown to reduce BP in selected patients (not illustrated).113-116 Creating an

arteriovenous fistula by implantation of an anastomosis device between the iliac artery and vein may reduce systemic vascular resistance and thereby reduces BP (Figure 4c).117,118

Importantly, most trials with innovative techniques are feasibility studies. Also it should be noted that the reported effects are mostly short-term (i.e. hours-days). The exciting positive open-label results create enthusiasm and quick developments lead to a rise in mainly small-scale, single centre studies. The Symplicity HTN-3 trial showed that sham-controlled as well as long-term follow-up confirmative studies are obligatory to further the role and learn the durability of the new technique and its long-term safety profile.

Renal sympathetic denervation

In the first decade of this century, a catheter based technique for resistant hypertension was developed disrupting renal sympathetic nerves without affecting other innervation (Figure 4d).119 Renal sympathetic nerve denervation (RDN) is achieved percutaneously via

the lumen of the renal artery, using a catheter connected to a radiofrequency generator. Renal sympathetic nerves in the artery wall are disrupted by applying a number of radio frequent ablations in a quadratic pattern up to 8 Watt, lasting for 120 seconds (Symplicity catheter). Since the proof of principle study in 2009 led to substantial BP reduction, RDN mushroomed as a treatment potential for resistant hypertension.120

Renal sympathetic nerve modification has been used long before pharmacological antihypertensive therapies were introduced. Radical surgical thoracic, abdominal and pelvic sympathetic denervation appeared a successful treatment for malignant hypertension. However, the high per-operative comorbidity, mortality and long-term complications replaced the technique by pharmacological agents.121 Observations from

bilateral nephrectomy in transplant patients that resulted in a decrease of systemic sympathetic nerve activity, lower renin activity and BP, suggests that native kidneys contribute to hypertension possibly by both afferent and efferent neural factors (Figure 3). These observations founded the pathophysiological rationale for RDN in non-transplant therapy resistant hypertensive patients.

RDN has been proven in non-kidney-transplant patients with therapy resistant hypertension to be safe, even for kidney function. In two studies (Symplicity HTN-1

(22)

and HTN-2), a pronounced offi ce based BP reduction of 22/11 to 32/12 mmHg was achieved after RDN, persisting for at least 6 months.120,122. These promising fi ndings

were countered by the Symplicity HTN-3 trial in which patients were randomized 2:1 to denervation or sham. The trial confi rmed feasibility and safety of RDN but failed to demonstrate effects on systolic BP.123 In non-sham studies, patient and

physician-related biases confounded in the positive results. Technical procedural and a large inter-individual variety in BP responses after RDN in Symplicity HTN-studies may be caused by incomplete nerve disruption.124

Nowadays, the initial widespread enthusiasm fuelled by Symplicity 1 and HTN-2 studies, has been tempered by the outcome of Symplicity HTN-3.125 At least the

Symplicity HTN trials have highlighted the importance of confounding in hypertension research, the trial effects, the importance of correctly diagnosing treatment resistant hypertension and the importance of medication adherence verifi cation. Native kidney denervation has a strong pathophysiological rationale in hypertensive renal transplant patients: it may decrease daily drug dose, thereby increasing adherence and following this one-time intervention and it is unlikely that the native kidney is harmed. Finally, the lowering of CVD risk can be of substantial benefi t. A feasibility study in renal transplant recipients was designed in 2012 (Netherlands Trial Registry number 3866, Academic Medical Center, Amsterdam).

Figure 4. Innovati ve strategies to lower blood pressure. Figure 4a: Electric baro-receptor

sti mulati on, Figure 4b: Mechanical baro-receptor sti mulati on, Figure 4c: Ileac arteriovenous fi stulati on, Figure 4d: Cathether-based renal sympatheti c denervati on.

4a. 4b.

(23)

Figure 5. 123I-mIBG pathway

Assessment of renal sympathetic nerve activity

To further our understanding of the pathogenesis of hypertension and the interplay of the kidney (either transplanted or not) and the autonomic nervous system, assessment of the human renal sympathetic nerves is essential. Assessment of renal sympathetic nerve activity in-vivo can only be performed using indirect methods. Norepinephrine spillover methods using radiotracer dilution is the gold standard method: regional venous sampling quantifies the norepinephrine released from the kidneys into the circulation. However, this method is invasive and the compound is not easily available in Europe. 123I-mIBG scintigraphy

We proposed the minimal-invasive renal 123Iodide-metaiodobenzylguanidine (123I-mIBG)

scintigraphy to measure sympathetic nerve activity. In nuclear cardiology, 123I-mIBG

is used to assess adrenergic integrity and functionality of nerves in myocardial tissue. Principally in heart failure patients, 123I-mIBG uptake and washout rates are

routinely used as prognostic factors.126-129

MIBG is a chemical modification of the false-neurotransmitter guanethidine (a former anti-hypertensive agent and potent neuron blocking agent). Uptake pathways for norepinephrine into the presynaptic terminals take up guanethidine into the presynaptic terminals. When mIBG is labelled to 123-I, visualization of sympathetic nerve terminals containing 123I-mIBG is possible (Figure

5). The amount of uptake of 123I-mIBG reflects density and functional intactness of

neural tissue within the organ, whereas washout, the rate at which the labelled mIBG is cleared from the synaptic cleft, reflects sympathetic activity. After heart transplantation, a progressive increase in myocardial uptake of 123I-mIBG is seen, proving reinnervation

of the myocardium.130 The changes in sympathetic nerve activity after RDN have been

(24)

OUTLINE OF THIS THESIS

Kidney transplant recipients are at a high risk for cardiovascular disease, for which hypertension is a prime modifiable risk factor.

Chapter 2 evaluates prevalence of hypertension and treatment efficacy in Dutch

kidney transplant recipients.

In Chapter 3, the epidemiology of hypertension after kidney transplantation is further

elaborated in paediatric and young adult kidney transplant recipients. It provides an analysis of the effects of transition from the paediatric to adult nephrologic care on hypertension control.

Since lifestyle interventions are thought a paramount antihypertensive therapy but have hardly been studied in kidney transplant recipients, dietary restriction of sodium chloride on blood pressure reduction in kidney transplant recipients is studied in Chapter 4.

The absence of nocturnal dipping in blood pressure, an additional risk factor for cardiovascular disease, may be related to the immunosuppressive regimens in kidney transplant recipients. Therefore, we evaluated the prolonged release vs. twice daily calcineurin inhibitor tacrolimus in Chapter 5.

Since the sympathetic nervous system contributes to the pathophysiology of hypertension and the rationale for renal catheter-based denervation, interruption of the nerves is a possible treatment target. In order to assess the changes in renal sympathetic nerve activity, we studied the nuclear imaging technique ¹²³I-mIBG scintigraphy in kidney transplant recipients.

In Chapter 6, this technique quantified reinnervation of renal sympathetic nerves in

kidney allografts at various time periods after transplantation.

In Chapter 7 we assessed the changes in renal sympathetic nerve activity after renal

denervation using ¹²³I-mIBG scintigraphy.

To conclude, we describa the case case of a kidney transplant recipient treated with renal denervation of his native kidneys for the treatment of resistant hypertension in

(25)

REFERENCES

1. ERA-EDTA Registry: Academic Medical Center. ERA-EDTA Registry Annual Report 2012. Amsterdam, The Netherlands, 2014: 78-79.

2. Segev DL. Innovative strategies in living donor kidney transplantation. Nat Rev Nephrol 2012; 8(6): 332-338.

3. Howard RJ, Patton PR, Reed AI et al. The changing causes of graft loss and death after kidney transplantation. Transplantation 2002;73: (12)

4. Ojo AO, Hanson JA, Wolfe RA, Leichtman AB, Agodoa LY, Port FK. Long-term survival in renal transplant recipients with graft function. Kidney Int 2000; 57(1): 307-313.

5. El-Zoghby ZM, Stegall MD, Lager DJ et al. Identifying Specific Causes of Kidney Allograft Loss. Am J Transplantation 2009; 9(3): 527-535.

6. National Institutes of Health. US Renal Data System (USRDS) 2012 Annual Data Report: Atlas of Chronic Kidney Disease and End-Stage Renal Disease in the United States: Chapter 7:Transplantation 2012: 283-294.

7. Ojo AO. Cardiovascular Complications After Renal Transplantation and Their Prevention. Transplantation 2006; 82(5): 603-611.

8. Parekh RS, Carroll CE, Wolfe RA, Port FK. Cardiovascular mortality in children and young adults with end-stage kidney disease. The Journal of Pediatrics 2002; 141(2): 191-197. 9. James PA, Oparil S, Carter BL. 2014 evidence-based guideline for the management of high

blood pressure in adults: Report from the panel members appointed to the eighth joint national committee (jnc 8). Journal of the American Medical Association 2014; 311(5): 507-520. 10. Lewington S, Clarke R, Qizilbash N, Peto R, Collins R, Prospective Studies C. Age-specific

relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 2002; 360(9349): 1903-1913.

11. Lim SS, Vos T, Flaxman AD et al. A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012; 380(9859): 2224-2260. 12. Sundstrom J, Arima H, Jackson R et al. Effects of blood pressure reduction in mild hypertension:

a systematic review and meta-analysis. Ann Intern Med 2015; 162(3): 184-191.

13. Law MR, Morris JK, Wald NJ. Use of blood pressure lowering drugs in the prevention of cardiovascular disease: meta-analysis of 147 randomised trials in the context of expectations from prospective epidemiological studies. Brit Med J 2009; 338: b1665.

14. KDIGO Clinical Practice Guideline for the Management of Blood Pressure in Chronic Kidney Disease 2012. Kidney International Supplements 2012; 2(5): 337-414.

15. Opelz G, Wujciak T, Ritz E, Collaborative-Transplant-Study. Association of chronic kidney graft failure with recipient blood pressure. Kidney Int 1998; 53(1): 217-222.

16. Kasiske BL, Anjum S, Shah R et al. Hypertension after kidney transplantation. American Journal of Kidney Diseases 2004; 43(6): 1071-1081.

17. Mange KC, Cizman B, Joffe M, Feldman HI. Arterial hypertension and renal allograft survival. J Am Med Assoc 2000; 283(5): 633-638.

18. Opelz G, Döhler B, Collaborative-Transplant-Study. Improved Long-Term Outcomes After Renal Transplantation Associated with Blood Pressure Control. Am J Transplant 2005; 5(11): 2725-2731.

19. Mancia G, Fagard R, Narkiewicz K et al. 2013 ESH/ESC Guidelines for the Management of Arterial Hypertension. the Task Force for the Management of Arterial Hypertension of the European Society of Hypertension (ESH) and of the European Society of Cardiology (ESC). J Hypertens 2013; 31(7): 1281-1357.

20. Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group Clinical Practice Guideline for the Care of Kidney Transplant Recipients. Am J Transplant 2009; 9(suppl 3): S1-S157.

21. Beluarte HJ, Gruskin A, Ingelfinger J, Stablein D, Tejani A. Analysis of hypertension in children post renal transplantation--a report of the North American Pediatric Renal Transplant Cooperative Study (NAPRTCS). Pediatric Nephrol 1994; 8(5): 570-573.

22. Calhoun DA, Jones D, Textor S et al. Resistant Hypertension: Diagnosis, Evaluation, and Treatment. Hypertension 2008; 51(6): 1403-1419.

(26)

23. de la Sierra A, Segura Jí, Banegas JR et al. Clinical Features of 8295 Patients With Resistant Hypertension Classified on the Basis of Ambulatory Blood Pressure Monitoring. Hypertension 2011; 57(5): 898-902.

24. Calhoun DA, Booth JN, 3rd, Oparil S et al. Refractory hypertension: determination of prevalence, risk factors, and comorbidities in a large, population-based cohort. Hypertension 2014; 63(3): 451-458.

25. Dobrowolski LC, Idu MM, Bemelman FJ. An unusual case of therapy-resistant hypertension. Kidney Int 2013; 84(6): 1290-1290.

26. Haydar AA, Covic A, Agharazii M, Jayawardene S, Taylor J, Goldsmith DJA. Systolic Blood Pressure Diurnal Variation is Not a Predictor of Renal Target Organ Damage in Kidney Transplant Recipients. Am J Transplant 2004; 4(2): 244-247.

27. Haydar AA, Covic A, Jayawardene S et al. Insights from ambulatory blood pressure monitoring: diagnosis of hypertension and diurnal blood pressure in renal transplant recipients. Transplantation 2004; 77(6): 849-853.

28. Kooman JP, Christiaans MHC, Boots JMM, van der Sande FM, Leunissen KML, van Hooff JP. A comparison between office and ambulatory blood pressure measurements in renal transplant patients with chronic transplant nephropathy. Am J Kidney Dis 2001; 37(6): 1170-1176. 29. Covic A, Segall L, Goldsmith DJA. Ambulatory blood pressure monitoring in renal

transplantation: should ABPM be routinely performed in renal transplant patients? Transplantation 2003; 76(11): 1640-1642.

30. Goldsmith DJA, Covic AC, Venning MC, Ackrill P. Ambulatory blood pressure monitoring in renal dialysis and transplant patients. Am J Kidney Dis 1997; 29(4): 593-600.

31. Hansen TW, Li Y, Boggia J, Thijs L, Richart T, Staessen JA. Predictive Role of the Nighttime Blood Pressure. Hypertension 2011; 57(1): 3-10.

32. Verdecchia P, Porcellati C, Schillaci G et al. Ambulatory blood pressure. An independent predictor of prognosis in essential hypertension. Hypertension 1994; 24(6): 793-801. 33. Amar J, Vernier I, Rossignol E et al. Nocturnal blood pressure and 24-hour pulse pressure are

potent indicators of mortality in hemodialysis patients. Kidney Int 2000; 57(6): 2485-2491. 34. Baumgart P, Walger P, Gerke M, Dorst KG, Vetter H, Rahn KH. Nocturnal hypertension in renal

failure, haemodialysis and after renal transplantation. J Hypertens Suppl 1989; 7(6): S70-S71. 35. Covic A, Gusbeth-Tatomir P, Mardare N, Buhaescu I, Goldsmith DJA. Dynamics of the

Circadian Blood Pressure Profiles after Renal Transplantation. Transplantation 2005; 80(9): 1168-1173.

36. Carpenter MA, Weir MR, Adey DB, House AA, Bostom AG, Kusek JW. Inadequacy of cardiovascular risk factor management in chronic kidney transplantation - evidence from the FAVORIT study. Clin Transplant2012; 26(4): E438-E446.

37. Vetromille F, Vincent P, Szwarc I, et al. Prevalence and Risk Factors of Noncontrolled and Resistant Arterial Hypertension in Renal Transplant Recipients. Transplantation 2015; 99(5): 1016-1022.

38. Kramer AM, van Stralen KJ, Jager KJ et al. Demographics of blood pressure and hypertension in children on renal replacement therapy in Europe. Kidney International 2011; 80(10): 1092-1098. 39. Seeman T. Ambulatory Blood Pressure Monitoring in Pediatric Renal Transplantation. Curr

Hypertens Rep 2012; 14(6): 608-618.

40. Cameron C, Vavilis G, Kowalski J, Tyden G, Berg UB, Krmar RT. An Observational Cohort Study of the Effect of Hypertension on the Loss of Renal Function in Pediatric Kidney Recipients. Am J Hypertens 2014; 27(4); 579-585.

41. Chue CD, Townend JN, Steeds RP, Ferro CJ. Arterial stiffness in chronic kidney disease: causes and consequences. Heart 2010; 96(11): 817-823.

42. Klassen PS, Lowrie EG, Reddan DN, et al. ASsociation between pulse pressure and mortality in patients undergoing maintenance hemodialysis. J Am Med Assoc 2002; 287(12): 1548-1555. 43. Bachelet-Rousseau C, Kearney-Schwartz A, Frimat L, Fay R, Kessler M, Benetos A. Evolution of

arterial stiffness after kidney transplantation. Nephrol Dial Transplant 2011; 26(10): 3386-3391. 44. Kim HS, Seung J, Lee JH, Chung BH, Yang CW. Clinical Significance of Pre-Transplant Arterial

Stiffness and the Impact of Kidney Transplantation on Arterial Stiffness. PLoS One 2015; 10(9): e0139138.

(27)

45. Grisk O, Klöting I, Exner J et al. Long-term arterial pressure in spontaneously hypertensive rats is set by the kidney. J Hypertens 2002; 20(1): 131-138.

46. Curtis JJ, Luke RG, Dustan HP et al. Remission of Essential Hypertension after Renal Transplantation. New Eng J Med 1983; 309(17): 1009-1015.

47. Guidi E, Cozzi MG, Minetti E, Bianchi G. Donor and recipient family histories of hypertension influence renal impairment and blood pressure during acute rejections. J Am Soc Nephrol 1998; 9(11): 2102-2107.

48. Guidi E, Menghetti D, Milani S, Montagnino G, Palazzi P, Bianchi G. Hypertension may be transplanted with the kidney in humans: a long-term historical prospective follow-up of recipients grafted with kidneys coming from donors with or without hypertension in their families. J Am Soc Nephrol 1996; 7(8): 1131-8.

49. Sarnak MJ, Levey AS, Schoolwerth AC et al. Kidney Disease as a Risk Factor for Development of Cardiovascular Disease: A Statement From the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation 2003; 108(17): 2154-2169.

50. Jardine AG. Assessing the relative risk of cardiovascular disease among renal transplant patients receiving tacrolimus or cyclosporine. Transplant Int 2005; 18(4): 379-384.

51. Baum M, Moe OW. Glucocorticoid-Mediated Hypertension: Does the Vascular Smooth Muscle Hold All the Answers? J Am Soc Nephrol 2008; 19(7): 1251-1253.

52. Saruta T. Mechanism of Glucocorticoid-Induced Hypertension. Hypertens Res 1996; 19(1): 1-8. 53. Withworth J. Mechanisms of glucocorticoid-induced hypertension. Kidney Int 1987; 31(5):

1213-1224.

54. Naesens M, Kuypers DRJ, Sarwal M. Calcineurin Inhibitor Nephrotoxicity. Clin J Am Soc Nephrol 2009; 4(2): 481-508.

55. Hortelano S, Castilla M, Torres AM, Tejedor A, Boscá L. Potentiation by Nitric Oxide of Cyclosporin A and FK506-Induced Apoptosis in Renal Proximal Tubule Cells. J Am Soc Nephrol 2000; 11(12): 2315-2323.

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

57. Hoorn EJ, Walsh SB, McCormick JA et al. The calcineurin inhibitor tacrolimus activates the renal sodium chloride cotransporter to cause hypertension. Nat Med 2011; 17(10): 1304-1309. 58. Hoorn EJ, Walsh SB, McCormick JA, Zietse R, Unwin RJ, Ellison DH. Pathogenesis of

calcineurin inhibitor-induced hypertension. J Nephrol 2012; 25(3): 269-275.

59. Ekberg H, Tedesco-Silva H, Demirbas A et al. Reduced Exposure to Calcineurin Inhibitors in Renal Transplantation. N Eng J Med 2007; 357(25): 2562-2575.

60. Meier-Kriesche HU, Ojo AO, Port FK, Arndorfer JA, Cibrik DM, Kaplan BRUC. Survival Improvement among Patients with End-Stage Renal Disease: Trends over Time for Transplant Recipients and Wait-Listed Patients. J Am Soc Nephrol 2001;12:(6):1293-1296.

61. DiBona GF, Kopp UC. Neural control of renal function. Physiol Rev 1997; 77(1): 75-197. 62. Klein IHHT, Ligtenberg G, Neumann J, Oey PL, Koomans HA, Blankestijn PJ. Sympathetic

Nerve Activity Is Inappropriately Increased in Chronic Renal Disease. J Am Soc Nephrol 2003; 14(12): 3239-3244.

63. Neumann J, Ligtenberg G, Klein II, Koomans HA, Blankestijn PJ. Sympathetic hyperactivity in chronic kidney disease: Pathogenesis, clinical relevance, and treatment. Kidney Int 2004; 65(5): 1568-1576.

64. Converse RL, Jacobsen TN, Toto RD et al. Sympathetic Overactivity in Patients with Chronic Renal Failure. New Eng J Med 1992; 327(27):1912-1918.

65. Hausberg M, Kosch M, Harmelink P et al. Sympathetic Nerve Activity in End-Stage Renal Disease. Circulation 2002; 106(15): 1974-1979. 66. Shannon JL, Headland R, MacIver AG, Ferryman SR, Barber PC, Howie AJ. Studies on the innervation of human renal allografts. J Pathol 1998; 186(1): 109-115. 67. Gazdar AF, Dammin GJ. Neural Degeneration and Regeneration in Human Renal Transplants. New Eng J Med 1970; 283(5): 222-224. 68. Kim J, Padanilam BJ. Renal denervation prevents long-term sequelae of ischemic renal injury. Kidney Int 2015; 87(2): 350-358.

(28)

69. Chadban SJ, Barraclough KA, Campbell SB et al. KHA-CARI guideline: KHA-CARI adaptation of the KDIGO Clinical Practice Guideline for the Care of Kidney Transplant Recipients. Nephrology 2012; 17(3): 204-214.

70. Curtis J, Diethelm A, Luke R, Whelchel J, Jones P. Benefits of removal of native kidneys in hypertension after renal transplantation. Lancet 1985; 2 (8458): 739-742.

71. Fricke L, Doehn C, Steinhoff J, Sack K, Jocham D, Fornara P. Treatment of Posttransplant Hypertension By Laparoscopic Bilateral Nephrectomy. Transplantation 1998; 65(9):1182-1187. 72. Vincenti F, Schena FP, Paraskevas S et al. A Randomized, Multicenter Study of Steroid

Avoidance, Early Steroid Withdrawal or Standard Steroid Therapy in Kidney Transplant Recipients. Am J Transplant 2008; 8(2): 307-316.

73. Pascual J, Zamora J, Galeano C, Royuela A, Quereda C. Steroid avoidance or withdrawal for kidney transplant recipients. Cochrane Database Syst Rev 2009: (1):CD005632.

74. Vincenti F, Jensik SC, Filo RS, Miller J, Pirsch J. A long-term comparison of tacrolimus (FK506) and cyclosporine in kidney transplantation: evidence for improved allograft survival at five years. Transplantation 2002; 73(5): 775-782.

75. Campistol JM, Romero R, Paul J, Gutierrez-Dalmau A. Epidemiology of arterial hypertension in renal transplant patients: changes over the last decade. Nephrol Dial Transplant 2004; 19(suppl 3): iii62-iii66.

76. Margreiter R. Efficacy and safety of tacrolimus compared with ciclosporin microemulsion in renal transplantation: a randomised multicentre study. Lancet 2002; 359(9308): 741-746. 77. Artz MA, Boots JMM, Ligtenberg G et al. Improved Cardiovascular Risk Profile and Renal

Function in Renal Transplant Patients after Randomized Conversion from Cyclosporine to Tacrolimus. J Am Soc Nephrol 2003; 14(7): 1880-1888.

78. Roodnat JI, Hilbrands LB, Hene RJ et al. 15-year follow-up of a multicenter, randomized, calcineurin inhibitor withdrawal study in kidney transplantation. Transplantation 2014; 98(1): 47-53.

79. Webber AB, Vincenti F. An Update on Calcineurin Inhibitor-Free Regimens: The Need Persists, but the Landscape has Changed. Transplantation 2016; 100(4): 836-43.

80. Chadban SJ, Barraclough KA, Campbell SB et al. KHA-CARI guideline: KHA-CARI adaptation of the KDIGO Clinical Practice Guideline for the Care of Kidney Transplant Recipients. Nephrology 2012;17(3): 204-14.

81. Chadban S, Chan M, Fry K et al. The CARI guidelines. Nutritional management of hypertension in adult kidney transplant recipients. Nephrology 2010; 15(Suppl 1): S56-S61.

82. Zelle DM, Kok T, Dontje ML et al. The role of diet and physical activity in post-transplant weight gain after renal transplantation. Clin Transplant 2013; 27(4): E484-E490.

83. Gore JL, Pham PT, Danovitch GM et al. Obesity and Outcome Following Renal Transplantation. Am J Transplant 2006; 6(2): 357-363.

84. Baum CL. Weight gain and cardiovascular risk after organ transplantation. J Parenter Enter Nutr 2001; 25(3): 114-9.

85. Zelle DM, Corpeleijn E, Stolk RP et al. Low physical activity and risk of cardiovascular and all-cause mortality in renal transplant recipients. Clin J Am Soc Nephrol 2011; 6(4): 898-905. 86. van den Berg E, Geleijnse JM, Brink EJ et al. Sodium intake and blood pressure in renal

transplant recipients. Nephrol Dial Transplant 2012; 27(8): 3352-9.

87. Prasad GV, Huang M, Nash MM, Zaltzman JS. Role of dietary salt intake in posttransplant hypertension with tacrolimus-based immunosuppression. Transplant Proc 2005; 37(4): 1896-1897. 88. Saint-Remy A, Somja M, Gellner K, Weekers L, Bonvoisin C, Krzesinski JM. Urinary and dietary

sodium and potassium associated with blood pressure control in treated hypertensive kidney transplant recipients: an observational study. BMC Nephrol 2012; 13(1): 121.

89. Cross NB, Webster AC, Masson P, O’Connell PJ, Craig JC. Antihypertensive treatment for kidney transplant recipients. Cochrane Database Syst Rev 2009; (3): CD003598.

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

91. Hoorn EJ, Walsh SB, McCormick JA, Zietse R, Unwin RJ, Ellison DH. Pathogenesis of calcineurin inhibitor-induced hypertension. J Nephrol 2012; 25(3): 269-275.

92. Wadei HM, Textor SC. Hypertension in the kidney transplant recipient. Transplant Rev 2010; 24(3): 105-120.

(29)

93. 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 2007; 7(10): 2350-2360.

94. Rosenberger J, Geckova AM, van Dijk JP et al. Prevalence and characteristics of noncompliant behaviour and its risk factors in kidney transplant recipients. Transpl Int 2005; 18(9): 1072-1078. 95. Dobbels F, Van Damme-Lombaert R, Vanhaecke J, De Geest S. Growing pains: non-adherence

with the immunosuppressive regimen in adolescent transplant recipients. Pediatr Transplant 2005; 9(3): 381-390.

96. Denhaerynck K, Dobbels F, Cleemput I et al. Prevalence, consequences, and determinants of nonadherence in adult renal transplant patients: a literature review. Transpl Int 2005; 18(10): 1121-1133.

97. Terebelo S, Markell M. Preferential adherence to immunosuppressive over nonimmunosuppressive medications in kidney transplant recipients. Transplant Proc 2010; 42(9): 3578-3585.

98. Shemesh E, Annunziato RA, Arnon R, Miloh T, Kerkar N. Adherence to medical recommendations and transition to adult services in pediatric transplant recipients. Curr Opin Organ Transplant 2010; 15(3): 288-292.

99. LaRosa C, Glah C, Baluarte HJ, Meyers KE. Solid-organ transplantation in childhood: transitioning to adult health care. Pediatrics 2011; 127(4): 742-753.

100. Watson AR. Non-compliance and transfer from paediatric to adult transplant unit. Pediatric Nephrol 2000; 14(6): 469-472.

101. Harden PN, Walsh G, Bandler N et al. Bridging the gap: an integrated paediatric to adult clinical service for young adults with kidney failure. Brit Med J 2012; 344:e3718.

102. Osterberg L, Blaschke T. Adherence to medication. N Engl J Med 2005; 353(5): 487-497. 103. Claxton AJ, Cramer J, Pierce C. A systematic review of the associations between dose

regimens and medication compliance. Clin Ther 2001; 23(8): 1296-1310.

104. Crowley R, Wolfe I, Lock K, McKee M. Improving the transition between paediatric and adult healthcare: a systematic review. Arch Dis Child 2011; 96(6): 548-553.

105. Schroeder K, Fahey T, Ebrahim S. Interventions for improving adherence to treatment in patients with high blood pressure in ambulatory settings. Cochrane Database Syst Rev 2004; (2): CD004804.

106. Paulis L, Rajkovicova R, Simko F. New developments in the pharmacological treatment of hypertension: dead-end or a glimmer at the horizon? Curr Hypertens Rep 2015; 17(6): 557. 107. Scheffers IJ, Kroon AA, Schmidli J et al. Novel baroreflex activation therapy in resistant

hypertension: results of a European multi-center feasibility study. J Am Coll Cardiol 2010; 56(15): 1254-1258.

108. Alnima T, Scheffers I, De Leeuw PW et al. Sustained acute voltage-dependent blood pressure decrease with prolonged carotid baroreflex activation in therapy-resistant hypertension. J Hypertens 2012; 30(8): 1665-1670.

109. Wallbach M, Lehnig LY, Schroer C et al. Effects of Baroreflex Activation Therapy on Ambulatory Blood Pressure in Patients With Resistant Hypertension. Hypertension 2016; 67(4): 701-709. 110. Habib N, Mahmoodi BK, Bos WJ et al. Initial experience with therapeutic geometric

modification of the carotid bulb for true resistant hypertension. Euro Int 2015; 11(1): 117-120. 111. Spiering W, Van Der Heyden J, Devireddy C, Foster MT, 3rd, Bates MC, Kroon AA. Controlling

and Lowering Blood Pressure with the Mobiushd Device: First-in-Man Results (Calm-Fim Study). J Hypertens 2015; 33(Suppl 1): e86.

112. Chapleau MW, Heesch CM, Abboud FM. Prevention or attenuation of baroreceptor resetting by pulsatility during elevated pressure. Hypertension 1987; 9(6 Pt 2): III137-141.

113. Frank H, Heusser K, Geiger H, Fahlbusch R, Naraghi R, Schobel HP. Temporary reduction of blood pressure and sympathetic nerve activity in hypertensive patients after microvascular decompression. Stroke 2009; 40(1): 47-51.

114. Sindou M. Is there a place for microsurgical vascular decompression of the brainstem for apparent essential blood hypertension? a review. Adv Tech Stand Neurosurg 2015; 42: 69-76. 115. Sindou M, Mahmoudi M, Brinzeu A. Hypertension of neurogenic origin: effect of microvascular

(30)

pressure in a prospective series of 48 patients with hemifacial spasm associated with essential hypertension. J Neurosurg 2015; 123(6): 1405-1413.

116. Legrady P, Voros E, Bajcsi D, Fejes I, Barzo P, Abraham G. Observations of changes of blood pressure before and after neurosurgical decompression in hypertensive patients with different types of neurovascular compression of brain stem. Kidney Blood Press Res 2013; 37(4-5): 451-457.

117. Lobo MD, Sobotka PA, Stanton A et al. Central arteriovenous anastomosis for the treatment of patients with uncontrolled hypertension (the ROX CONTROL HTN study): a randomised controlled trial. Lancet 2015; 385(9978): 1634-41.

118. Fudim M, Stanton A, Sobotka PA, Dolan E, Krum H. The thrill of success: central arterial-venous anastomosis for hypertension. Curr Hypertens Rep 2014; 16(12): 497.

119. Schlaich MP, Sobotka PA, Krum H, Lambert E, Esler MD. Renal sympathetic-nerve ablation for uncontrolled hypertension. N Engl J Med 2009; 361(9): 932-934.

120. Krum H, Schlaich M, Whitbourn R et al. Catheter-based renal sympathetic denervation for resistant hypertension: a multicentre safety and proof-of-principle cohort study. Lancet 2009; 373(9671): 1275-1281.

121. Morrissey DM, Brookes VS, Cooke WT. Sympathectomy in the treatment of hypertension; review of 122 cases. Lancet 1953; 1(6757): 403-408.

122. Symplicity HTNI, Esler MD, Krum H et al. Renal sympathetic denervation in patients with treatment-resistant hypertension (The Symplicity HTN-2 Trial): a randomised controlled trial. Lancet 2010; 376(9756): 1903-1909.

123. Bhatt DL, Kandzari DE, O’Neill WW et al. A controlled trial of renal denervation for resistant hypertension. N Engl J Med 2014; 370(15): 1393-1401.

124. Vink EE, Goldschmeding R, Vink A, Weggemans C, Bleijs RL, Blankestijn PJ. Limited destruction of renal nerves after catheter-based renal denervation: results of a human case study. Nephrol Dial Transplant 2014; 29(8): 1608-1610.

125. Yao Y, Zhang D, Qian J, Deng S, Huang Y, Huang J. The effect of renal denervation on resistant hypertension: Meta-analysis of randomized controlled clinical trials. Clin Exp Hypertens 2016; 38(3): 278-286. 126. Jacobson AF, Senior R, Cerqueira MD et al. Myocardial iodine-123 meta-iodobenzylguanidine imaging and cardiac events in heart failure. Results of the prospective ADMIRE-HF (AdreView Myocardial Imaging for Risk Evaluation in Heart Failure) study. J Am Coll Cardiol 2010; 55(20): 2212-2221.

127. Flotats A, Carrio I, Agostini D et al. Proposal for standardization of 123I-meta-iodobenzylguanidine (MIBG) cardiac sympathetic imaging by the EANM Cardiovascular Committee and the European Council of Nuclear Cardiology. Eur J Nucl Med Mol Imaging 2010; 37(9): 1802-1812.

128. Somsen GA, Verberne HJ, Fleury E, Righetti A. Normal values and within-subject variability of cardiac I-123 MIBG scintigraphy in healthy individuals: implications for clinical studies. J Nucl Cardiol 2004; 11(2): 126-33.

129. Verberne HJ, Brewster LM, Somsen GA, van Eck-Smit BL. Prognostic value of myocardial 123I-metaiodobenzylguanidine (MIBG) parameters in patients with heart failure: a systematic review. Eur Heart J 2008; 29(9): 1147-59.

130. Buendia-Fuentes F, Almenar L, Ruiz C et al. Sympathetic reinnervation 1 year after heart transplantation, assessed using iodine-123 metaiodobenzylguanidine imaging. Transplant Proc 2011; 43(6): 2247-2248.

131. van Brussel PM, Eeftinck Schattenkerk DW, Dobrowolski LC et al. Effects of Renal Sympathetic Denervation on Cardiac Sympathetic Activity and Function in Patients with Therapy Resistant Hypertension. Int J Cardiol 2015; 202: 609-614.

(31)
(32)

Linn C. Dobrowolski, Frederike J. Bemelman,

Karlijn A.M.I. van der Pant, Andries J. Hoitsma,

Ineke J.M. ten Berge, C.T. Paul Krediet

CHAPTER 2

TREATMENT EFFICACY OF HYPERTENSION

IN KIDNEY TRANSPLANT RECIPIENTS

IN THE NETHERLANDS

(33)

ABSTRACT Background

Hypertension in kidney transplant recipients jeopardises graft and patient survival. Guidelines suggest blood pressure targets of ≤ 130/80 mmHg and sodium intake < 90 mmol/day.

Methods

Since the efficacy of antihypertensive treatment among kidney transplant recipients is unknown, we analysed data on office-based blood pressure and use of antihypertensive drugs from the Netherlands Organ Transplant Registry on 5415 kidney transplant recipients. Additionally, we studied dosages, prevalence of treatment-resistant hypertension and 24 hour sodium excretion in 534 kidney transplant recipients from our centre to explore possibilities for therapy optimisation.

Results

In patients registered in the Netherlands Organ Transplant Registry, median blood pressure was 134/80 mmHg (interquartile range 122-145/70-85). In 77.2%, the blood pressure was ≥ 130/80 mmHg; of these patients 10.4% had no registered use, 30.0% used one and 25.9% used ≥ 3 classes of antihypertensive agents. Parameters from our centre were comparable: 78.7% had a median blood pressure of ≥ 130/80 mmHg of whom 14.5% had no registered use of antihypertensives and 26.4% used ≥ 3 classes. Sub-maximal dosages were prescribed in 74.0% of the kidney transplant recipients with a blood pressure of ≥ 130/80 mmHg while using at least one antihypertensive agent. Treatment-resistant hypertension was present in 7.7%. Median 24 h sodium excretion was 147 mmol/day (interquartile range 109-195).

Conclusions

This study suggests that therapeutic optimisation of antihypertensive treatment in kidney transplant recipients is, in theory, frequently possible by intensifying pharmacological treatment and by providing more advice on dietary sodium restrictions.

(34)

INTRODUCTION

Although kidney transplantation is the superior treatment for end-stage renal disease, kidney transplant recipients continue to have a high risk for cardiovascular morbidity and mortality. The annual risk for cardiovascular death is about 50-fold as compared with the general population and cardiovascular disease is the leading cause of morbidity and mortality in kidney transplant recipients.1-3

Hypertension is the foremost modifiable medical risk factor for cardiovascular disease in kidney disease. In addition, hypertension jeopardises renal allograft function, leading to graft loss.4-7 Various studies, dating from before 2009, indicated that hypertension

amongst kidney transplant recipients was prevalent in up to > 90%.4,8,9 Authoritative

guidelines recommend a target blood pressure of < 130/80 mmHg in kidney transplant recipients.7,10 The efficacy of antihypertensive treatment in kidney transplant recipients

has not been studied since. Against this background, we set out to study the efficacy of the current treatment of hypertension in kidney transplant recipients and to assess the number and dosages of prescribed antihypertensive drugs. Since sodium intake is a recognised determinant of blood pressure and sodium restriction is a major therapeutic antihypertensive intervention, we also surveyed the dietary sodium intake.

METHODS

We performed two separate, retrospective cross-sectional analyses: i.e. on data retrieved from the Netherlands Organ Transplant Registry (NOTR) and from the clinical files of kidney transplant recipients at our own institution, respectively.

Netherlands Organ Transplant Registry

The NOTR registry is a nationwide registry of kidney transplant recipients from the eight kidney transplant centres in the Netherlands, including our institution. The NOTR registry is managed by the Dutch Transplant Foundation and includes patient and donor characteristics and a variety of clinical parameters, such as office blood pressure and prevalent medications. In the first year after transplantation, registry follow-up is at month 3, thereafter on a yearly basis. We retrieved data on patient characteristics, kidney graft function, office-based systolic and diastolic blood pressure and the number of classes of antihypertensive drugs in the patients registered on 31 December 2011. For patients within the first year after transplantation, we included data from the latest visit. Table 1 summarises these variables.

Local data

To provide additional information about determinants of hypertension that could not be retrieved from the NOTR, we performed a retrospective survey on the medical files of the prevalent kidney transplant recipients at our kidney transplant centre in Amsterdam in September 2012. On average these patients visit the outpatient clinic

(35)

four times per year. We collected data on patient characteristics including ethnicity, kidney graft function, office-based systolic and diastolic blood pressure and prevalent classes of antihypertensive and immunosuppressive drugs and their dosages. In all patients 24-hour urine collections are routinely performed at each outpatient clinic visit. There we were able to assess daily sodium excretion as a proxy of dietary intake parallel to the blood pressure readings. Urine sodium excretion was measured at least four weeks after adjustment or initiation of diuretic treatment. Therefore these measurements represented a steady state in which sodium intake and excretion were equal. We defined the possibility for optimisation of antihypertensive treatment as the option to initiate antihypertensive treatment or the option to increase the number and/ or dosage of the prescribed antihypertensive agents up to the maximum recommended dosage in our local protocol (Table 2).

Statistical analysis

All data were included in a master file and statistical analyses were performed using SigmaStat (Jandel Scientific Software, San Jose, California USA). Normally distributed data are represented as mean and SD; non-normally distributed data as median and interquartile ranges. Under Dutch law this retrospective, descriptive study was exempt from medical ethics review.

RESULTS

Netherlands Organ Transplant Registry data

On 31 December 2011, 5770 patients of 18 years and older were registered in the NOTR as living with a functioning kidney transplant. Recent blood pressure measurements were missing in 355 patients (6.2%), who were excluded from further analysis. Median age was 48 years (interquartile range (IQR) 36-58) and time after transplantation 5.0 years (IQR 2-11). Median plasma creatinine was 126 μmol/l (IQR 101-163) and proteinuria was 0.11 g/l (IQR 0.03-0.30). Most patients were treated with a calcineurin inhibitor (CNI). Tacrolimus was prescribed in 58.2% and cyclosporine in 36.9% of the patients. Prednisolone was prescribed in 89.5% of the patients, mostly in combination with a CNI. Mycophenolate mofetil (MMF) was used in addition to the CNI and/or prednisolone regimen in 73.8%. Azathioprine was given to 5.3% of the kidney transplant patients and a mammalian target of rapamycin (mTOR) inhibitor to 5.1%. Median blood pressure was 134/80 mmHg (IQR 122-145/70-85). These data are summarised in Table 1 and Figure 1. The examination of the numbers of classes of antihypertensive drugs prescribed showed that at least one class of blood pressure lowering agents was prescribed in 87.8% of the patients. Of all kidney transplant recipients with a blood pressure ≥ 130/80 mmHg, 10.4% had no prescription for any antihypertensive drug, 30.0% used one antihypertensive agent, 33.7% used two and 25.9% used three or more different classes of antihypertensive drugs (Figure 2).

(36)

Local data

Patient characteristics are shown in Table 1. Data on n=539 prevalent patients living with a functioning kidney transplant on 1 September 2012 were included. There were missing data on recent blood pressure in five patients. Therefore, we further analysed the data on 534 patients. There were nine patients who had missing data on 24-hour urine specimens. In this cohort, the median age of 54.5 years (IQR 43-64) was slightly higher and time after transplantation of 4.4 years (IQR 1.3-9.7) was shorter than in the NOTR survey. Ethnic diversity was broad with 76% being Caucasian and of whom 64% were of Dutch descent. Median plasma creatinine was 143 μmol/l (IQR 112-183) and proteinuria was 0.09 g/l (IQR 0.06-0.18). The CNI tacrolimus was prescribed in 53.4% of the patients, with median dosages of 6 mg/day (IQR 4-8) and with associated plasma trough levels of 7.4 μg/l (IQR 5.5-9.2). Cyclosporine was prescribed in 20.8%, and 93.6% of the kidney transplant recipients were treated with prednisolone with median daily dosages of 10 mg (5-10 mg).

In our centre, 58.1% of our kidney transplant recipients received MMF, 14.0% azathioprine and 4.1% an mTOR inhibitor.

Kidney transplant recipients had a median blood pressure of 134/81 mmHg (124- 146/76-88). In 420 patients (78.6%) blood pressure was ≥ 130/80 mmHg, of whom 14.5% were not taking an antihypertensive drug, 29.3% used one, 29.8% two and 26.8% used three or more different classes of blood pressure lowering agents. Blood pressure ≥ 140/90 mmHg was found in 43.8% of our patients. Of the 420 patients with blood pressure ≥ 130/80 mmHg, 24.8% were taking three or more antihypertensive

Figure 1. Blood pressure of kidney transplant recipients (data from the NOTR). Blood pressure

is classified into three categories (≤ 129/79 mmHg, between 130/80 and 139/89 mmHg and ≥ 140/90 mmHg).

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

phase of the disease focuses on conserved epitopes, is associ- ated with viral clearance, the absence of the described epitope- specific CD4 1 T-cell response in patients developing

Using the Fredholm alternative for the existence of bounded solutions of a linear variational problem, we have derived in this paper, a Mel’nikov vector for

These were sampled along the principal environmental gradients in a wide rain forest area in Colombian Amazonia , and adjacent (Amazon) areas of Ecuador and

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of

Stedets ånd: Dan Ringgaard tager sine læsere med på en essayistisk lystvandring uden livrem og seler [Review of: D. Ringgaard

The aim of this study was to assess the detection rate and risk of CIN and cervical cancer in women with IBD as compared with the general Dutch female population, and to assess

The distribution in time and space; the shape of the pits; and the pit contents (including fire-cracked flint, charred logs, charred wetland plants, multiple wood species in