Introduction to Renal Therapeutics

Introduction to Renal Therapeutics

Edited by Caroline Ashley MSc, BPharm (Hons), MRPharmS Lead Specialist Pharmacist, Renal Services Royal Free Hospital Royal Free Hampstead NHS Trust London, UK Clare Morlidge BPharm (Hons), DipClinPharm, MRPharmS Cardiothoracic Directorate Pharmacist Lister Hospital East and North Hertfordshire NHS Trust Hertfordshire, UK

On behalf of the UK Renal Pharmacy Group

London • Chicago Published by the Pharmaceutical Press An imprint of RPS Publishing

1 Lambeth High Street, London SE1 7JN, UK 100 South Atkinson Road, Suite 200, Grayslake, IL 60030–7820, USA

© Pharmaceutical Press 2008

is a trade mark of RPS Publishing

RPS Publishing is the publishing organisation of the Royal Pharmaceutical Society of Great Britain

First published 2008

Typeset by Photoprint, Torquay, Devon Printed in Great Britain by Cambridge University Press, Cambridge

ISBN 978 0 85369 688 9

All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, without the prior written permission of the copyright holder. The publisher makes no representation, express or implied, with regard to the accuracy of the information contained in this book and cannot accept any legal responsibility or liability for any errors or omissions that may be made. The right of Caroline Ashley and Clare Morlidge to be identified as the authors of this work has been asserted by them in accordance with the Copyright, Designs and Patents Act, 1988.

A catalogue record for this book is available from the British Library Contents

Preface xi About the editors xiii Contributors xv Abbreviations xvii

1 What are the functions of the kidney? 1 The functional anatomy of the kidney 1 Other functions of the kidneys 5 Conclusion 7

2 Laboratory tests and investigations 9 Standard tests and investigations for the general adult patient population 9 Haematological tests and investigations 13 Urine tests 14 Renal biopsy 15 Immunological tests 15 Serum and urine electrophoresis 15 Radiological tests 16 Describing and measuring renal function 16 References 18

3 Acute renal failure 21 Definition 21 Incidence 21 Clinical features 22 Causes 22 Symptoms/signs 27 Distinguishing acute from chronic renal impairment 28

v vi Contents

Management 28 Additional points 31 Outcome 31 Conclusion 31 References 33 Further reading 34

4 Chronic renal failure 35 Definition 35 Clinical features 35 Incidence 37 Causes 37 Symptoms and consequences 38 Influence of chronic renal disease on drug handling 41 Renal replacement therapies 42 Conclusion 42 References 43 Further reading 44

5 Renal anaemia 45 Maintenance of serum haemoglobin in the healthy individual 45 The development and consequences of anaemia of chronic kidney disease 47 Assessing the anaemia of chronic kidney disease 49 Iron and erythropoiesis-stimulating agents available for the correction of renal anaemia 51 Iron preparations 53 Initiating iron and erythropoiesis-stimulating agents therapy 53 Failure to respond to therapy 54 Summary 54 References 55

6 Renal bone disease 57 Pathophysiology 57 Complications for the skeleton and progression of renal bone disease 60 Treatment with phosphate binders 63 Treatment with vitamin D 65 The role of parathyroidectomy 66 Contents vii

Therapeutic targets 68 New management strategies and new therapeutic agents for renal bone disease 69 References 74 Further reading 74

7 Hypertension and hyperlipidaemia 77 Hypertension 77 Hyperlipidaemia 81 References 84

8 Renal replacement therapy 85 Haemodialysis 85 Continuous renal replacement therapies 91 Haemodiafiltration 95 Peritoneal dialysis 96 References 105

9 Renal transplantation 107 The donor organ 107 Transplant rejection and its prevention 108 Transplant immunosuppression 109 Post-transplant complications 117 The pharmacist’s role in caring for transplant patients 121 References 124

10 Drug dosing in patients with renal impairment and during renal replacement therapy 127 Drug handling considerations in patients with renal impairment 127 Assessment of renal function 129 Adjusting doses for patients with renal impairment 132 Drug handling considerations in patients undergoing renal replacement therapy 133 Adjusting drug doses for patients undergoing renal replacement therapy 135 Summary 136 References 137 Further reading 137 viii Contents

11 Drug-induced kidney disease 139 Pre-renal (or haemodynamic) acute renal failure 139 Intrinsic renal toxicity 141 Post-renal damage (obstructive uropathy) 142 Conclusion 143 References 144

12 Autoimmune kidney disease 145 Pathogenesis 145 Clinical manifestations 145 Principles of treatment 147 Antineutrophil cytoplasmic autoantibody (ANCA)-associated vasculitis 148 Goodpasture’s syndrome 153 Systemic lupus erythematosus 154 IgA nephropathy 156 Membranous glomerulonephritis 158 Membranoproliferative glomerulonephritis 159 Cryoglobulinaemic glomerulonephritis 159 Conclusions 160 References 161

13 Paediatric kidney disease 163 Creatinine and glomerular filtration rate 163 Monitoring and treatment of children with chronic renal failure 164 Transplantation 169 Dialysis 173 Drug prescribing in renal disease 178 References 179 Further reading 180

14 Renal pharmacy in critical care 181 Preventing acute renal failure 181 Renal replacement therapy in the intensive care unit 182 Drug therapies specific to critical care areas 184 General comments on administering drugs to patients with renal impairment 188 References 189 Contents ix

15 Pain control in renal impairment 191 Assessing pain 191 The analgesic ladder 191 Analgesics and renal impairment 191 Individual agents 192 References 201 Further reading 204

16 Diabetes management in kidney disease 205 Definitions of diabetic renal disease 205 The actual structural changes that occur in the diabetic kidney 206 The diagnosis of diabetic renal disease 206 The prevention of diabetic renal disease 206 The screening and referral process for diabetic renal disease 207 The management of diabetes in kidney disease 207 References 216

17 Myeloma and kidney disease 217 Clinical presentation of multiple myeloma 218 Investigation and diagnosis of myeloma 218 Myeloma kidney 218 Treatment options 219 Summary 223 References 224

18 Palliative care 225 Prescribing for symptom control in patients with stage 5 chronic kidney disease managed without dialysis 226 Prescribing in the last few days of life 233 Conclusions 235 References 237

19 Dietary management of kidney disease 239 Historic pre-dialysis dietary intervention 239 Pre-dialysis dietary management 239 Dietary management of renal replacement therapy 244 Vitamin and minerals 247 x Contents

Malnutrition 248 References 252

20 Travelling and vaccines 253 Planning holidays 253 General travel health advice 254 Travelling abroad with medication 257 Malaria 259 Travel vaccinations 261 Conclusion 263 References 264 Further information 265

Case study answers 267 Glossary 289 Index 295 Preface

Welcome to the first edition of the Introduction book is to help pharmacy students, pharmacists to Renal Therapeutics. In 1995, the UK Renal and other healthcare workers to understand and Pharmacy Group recognised that there was a promote the safe and effective use of drug ther- lack of readily accessible information for phar- apies in renal failure. macists, either with an interest in renal medi- The case studies and questions have been cine, or newly appointed as specialist kept separate from the answers in order to practitioners in the renal field. As a result, mem- encourage readers to formulate their own bers of the RPG wrote and produced the answers before reading the author’s. The Beginner’s Guide to Renal Pharmacy. It was answers section illustrates how the questions designed to be a comprehensive introductory should be approached and what factors need to guide to the causes, effects and management of be taken into consideration when formulating a renal failure, and as such, a copy has since been response. The answers are based on clinical given to each new member of the RPG. In addi- opinion at the time of writing, but they also tion, the Beginner’s Guide has become required represent, to some degree, the opinions of the reading for some post-graduate pharmacy authors themselves. As time passes and new courses. drugs and new information become available, The Beginner’s Guide has now been updated some readers may disagree with the decisions and expanded to become the Introduction to arrived at by the authors, which is entirely Renal Therapeutics. This guide has been written appropriate. by practising renal pharmacists and other We would like to take this opportunity to healthcare professionals who work in renal thank all those pharmacists, dieticians and units. The mix of authors and the depth of their nurses who have contributed chapters for this ‘hands on’ experience has been used to provide book. Writing these chapters requires an enor- a text that directs the reader to the important mous amount of time and effort, and everyone factors in renal therapies. It highlights points has given unstintingly of both. We hope that that may not be readily accessible in any other this book will be of use to all those pharmacists single text, but still bears in mind the practi- who aspire to become a specialist renal phar- tioner new to renal pharmacy. macist, as well as those who just wish to This book is not meant to be exclusive to improve their renal knowledge. pharmacists working in renal units, but should be used by all those who wish to familiarise Caroline Ashley themselves with the issues concerning drug Clare Morlidge therapy for the renally impaired. The aim of this July 2007

xi

About the editors

Clare Morlidge BPharm (Hons), DipClinPharm, Hospital in London. She is the Chair of the UK MRPharmS graduated from Bradford University, Renal Pharmacy Group, has served on the and undertook the clinical pharmacy diploma Council of the British Renal Society, and was whilst working as a basic grade pharmacist in involved with the development of the Renal Worcester Hospital. Clare moved to Walsgrave National Service Framework. Hospital, Coventry where she worked as a renal Caroline studied pharmacy at the School of pharmacist for a number of years. During this Pharmacy, University of London, and later time Clare wrote articles in the Pharmaceutical gained an MSc in Clinical Pharmacology from Journal, Nephrology Dialysis and Transplantation the University of East London. Her interest in and British Journal of Renal Medicine. She was also renal medicine began during her time as a basic involved in writing the renal unit’s Help I’ve got grade pharmacist at Guy’s Hospital, and she has Renal Failure book, Dr Andy Stein’s Kidney Dialysis been specialist renal pharmacist at the Royal and Transplantation, and was on the editorial Free since 1991. She is co-editor of the Renal board for the Renal Pharmacy Group’s Renal Drug Drug Handbook and has written numerous arti- Handbook 2004 edition. cles for various pharmacy journals, including Clare currently works at the Lister Hospital in the Pharmaceutical Journal and Hospital Stevenage as a senior cardiothoracic pharmacist. Pharmacist. In addition, she has contributed chapters to several books, including Drugs in Caroline A Ashley MSc, BPharm (Hons), Use, and the forthcoming edition of the Oxford MRPharmS has worked as a hospital pharmacist Textbook of Medicine. She has recently been for 20 years, and is currently the lead specialist invited to join the editorial board of the British pharmacist for renal services at the Royal Free Journal of Renal Medicine.

xiii

Contributors

Caroline Ashley MSc, BPharm (Hons), Diane Green BSc (Hons), SRD MRPharmS Dietetic Manager, Hope Hospital, Salford Lead Specialist Pharmacist, Renal Services, Royal PCT Free Hospital Hampstead NHS Trust, London Elizabeth Lamerton BSc, DipClinPharm, Stephen Ashmore MPhil, BPharm, MRPharmS MRPharmS Clinical Pharmacy Team Leader, Acute Medicine/ Senior Clinical Pharmacist, Renal Medicine, Renal Medicine, Leeds General Infirmary, Leeds Salford Royal NHS Foundation Trust Teaching Hospitals NHS Trust Mark Lee BSc (Hons), MRPharmS Robert Bradley MSc, BPharm, MRPharmS Advanced Level Pharmacist, Renal Transplant, Lead Pharmacist for Nephrology and Renal Leeds Teaching Hospitals NHS Trust Transplantation, University Hospital of Wales, Cardiff & Vale NHS Trust Anne Millsop BSc (Hons), DipClinPharm, Cert Clin Pharm Teach, MRPharmS Aileen Currie BSc (Hons), MRPharmS Renal Pharmacist, Oxford Transplant Centre, Renal Pharmacist, Crosshouse Hospital, Oxford Radcliffe Hospitals NHS Trust Kilmarnock Clare Morlidge BPharm (Hons), DipClin- John Dade BPharm, MPharm, MRPharmS Pharm, MRPharmS Clinical Pharmacist – Critical Care, St James’s Cardiothoracic Pharmacist, Lister Hospital, University Hospital, Leeds Teaching Hospitals Stevenage NHS Trust Emma Murphy RN, DipNurStudies Andrea Devaney BPharm, DipClinPharm, Clinical Nurse Specialist – Renal Palliative Care MRPharmS – Modernisation Initiative, Guy’s and St Renal Pharmacy Team Manager, Oxford Thomas’ NHS Foundation Trust, London Transplant Centre, Oxford Radcliffe Hospitals NHS Trust Fliss Murtagh EM, MRCGP, MSc Clinical Research Training Fellow, Dept of James Dunleavy BSc, MSc, MRPharmS Palliative Care, Policy & Rehabilitation, King’s Senior Clinical Pharmacist – Renal Services, College London Monklands Hospital, Airdrie, Lanarkshire Mrudula Patel MSc, BPharm, DipClinPharm Roger Fernandes MSc, BPharm, MRPharmS Pract, MRPharmS Deputy Chief Pharmacist, Mayday University Renal Directorate Pharmacist, Churchill Hos- Hospital, Croydon pital, Oxford

xv xvi Contributors

Susan Patey BSc (Pharmacy), MRPharmS Zoe Thain BSc, DipClinPharm, MRPharmS Lead Pharmacist, Nephrology, Great Ormond Senior Pharmacist, Renal Services, Lancashire Street Hospital for Children NHS Trust London Teaching Hospitals NHS Trust

Jane Pearson BSc (Hons), DipClinPharm, Marc Vincent BPharm, DipClinPharm, MRPharmS MRPharmS Senior Pharmacist Renal Medicine, Royal Lead Pharmacist Renal Services, Manchester Infirmary of Edinburgh Royal Infirmary

John Sexton, MSc, BPharm (Hons), MCPP, Hayley Wells MSc, BPharm, MRPharmS MRPharmS Senior Clinical Pharmacist, Renal Services, Guy’s Principal Pharmacist Lecturer-Practitioner, Royal and St. Thomas’ NHS Foundation Trust, London Liverpool and Broadgreen University Hospitals NHS Trust & Liverpool John Moores University Abbreviations

ACE angiotensin-converting enzyme ACR albumin:creatinine ratio ACTactivated clotting time ADH antidiuretic hormone ADPKD autosomal dominant polycystic disease AERD aspirin-exacerbated respiratory disease AGE advanced glycation end-products AIN acute interstitial nephritis AMR antibody-mediated rejection ANA antinuclear antibodies ANCA antineutrophil cytoplasmic antibodies APA anti-phospholipid antibodies APC antigen-presenting cells APD automated peritoneal dialysis APTT activated partial thromboplastin time ARF acute renal failure AT-II angiotensin II ATG antithymocyte globulin ATN acute tubular necrosis ATP adenosine triphosphate AUC area under the curve BP blood pressure BSA body surface area BUN blood urea nitrogen CAN chronic allograft nephropathy CAPD continuous ambulatory peritoneal dialysis CAVH/CAVHF continuous arterio-venous haemofiltration CAVHD continuous arterio-venous haemodialysis CAVHDF continuous arterio-venous haemodiafiltration CCPD continuous cycling peritoneal dialysis CHD coronary heart disease CKD chronic kidney disease CMV cytomegalovirus CNI calcineurin inhibitor CNS central nervous system COX cyclo-oxygenase CrCl creatinine clearance

xvii xviii Abbreviations

CRF chronic renal failure CRP C-reactive protein CRRTcontinuous renal replacement therapy CSS Churg–Strauss syndrome CTcomputed tomography CVP central venous pressure CVS cerebrovascular system CVVH/CVVHF continuous veno-venous haemofiltration CVVHDF continuous veno-venous haemodiafiltration DGF delayed graft function DIC disseminated intravascular coagulation DNA deoxyribonucleic acid DSA donor specific antibodies DTPA diethylenetriamine penta-acetic acid DVTdeep vein thrombosis EABV effective arteriolar blood volume EBCTelectron beam computed tomography EBPG European Best Practice guidelines EBV Epstein-Barr virus ECG electrocardiogram ECHO echocardiogram EDTA ethylenediamine tetra-acetic acid eGFR estimated glomerular filtration rate EHIC European Health Insurance Card ESA erythropoiesis-stimulating agent ESR erythrocyte sedimentation rate ESRD end stage renal disease ESRF end stage renal failure GBM glomerular basement membrane GFR glomerular filtration rate GI gastrointestinal GLP-1 glucagon-like peptide 1 H3G hydromorphone-3-glucuronide HD intermittent haemodialysis HDU high dependency unit HES hydroxyethyl starch HITheparin-induced thrombocytopenia HIV human immunodeficiency virus HLA human leukocyte antigens HMG-CoA 3-hydroxy-3-methylglutaryl coenzyme A HPThyperparathyroidism HRThormone replacement therapy HSP Henoch–Schönlein purpura HUS haemolytic uraemic syndrome IBW ideal body weight ICU intensive care unit IDDM insulin-dependent diabetes mellitus IDH ischaemic heart disease IDPN intradialytic parenteral nutrition IL-2 interleukin-2 Abbreviations xix

IM intramuscular INR international normalised ratio IP intraperitoneally iu international units IV intravenous IVP intravenous pyelogram IVU intravenous excretory urogram KCTT kaolin cephalin clotting time LFTliver function test LMWH low molecular weight heparin LVF left ventricular failure LVH left ventricular hypertrophy M3G morphine-3-glucuronide M6G morphine-6-glucuronide MAM monoacetylmorphine MDRD modified diet in renal disease MHC major histocompatibility complex MHRA Medicines and Healthcare Products Regulatory Agency MI myocardial infarction MMF mycophenolate mofetil MPA mycophenolic acid OR microscopic angitis MPO myeloperoxidase M/R modified release MRI magnetic resonance imaging MRSA methicillin-resistant Staphylococcus aureus MTOR mammalian target of rapamycin mw molecular weight NICE National Institute for Health and Clinical Excellence NIDDM non-insulin-dependent diabetes mellitus NKF-K/DOQU National Kidney Federation Dialysis Outcomes Quality Initiative NMDA N-methyl-D-aspartate NNH number needed to harm NNTnumber needed to treat NSAID non-steroidal anti-inflammatory drug OD once daily OHA oral hypoglycaemic agent OTC over the counter (medicine) PCA patient-controlled analgesia PCP Pneumocystis carinii pneumonia (now Pneumocystis jiroveci) PCR protein-creatinine ratio PD peritoneal dialysis PDF peritoneal dialysis fluid PE pulmonary embolism PETperitoneal equilibrium test PKD polycystic kidney disease PR3 proteinase-3 PRA panel reactive antibodies PRCA pure red cell aplasia PTH parathyroid hormone PTFE polytetrafluoroethylene xx Abbreviations

PTLD post-transplant lymphoproliferative disease RAS renal artery stenosis RBC red blood cells RNA ribonucleic acid RNI recommended nutrient intake ROD renal osteodystrophy RPGN rapidly progressive glomerulonephritis RRTrenal replacement therapy SC subcutaneous SCUF slow continuous ultrafiltration SIRS systemic inflammatory response syndrome SLE systemic lupus erythematosus SPC summary of product characteristics SPF sun protection factor SSRI selective serotonin reuptake inhibitor SUN serum urea nitrogen t1/2 elimination half life TB tuberculosis TCA tricyclic antidepressant TDM therapeutic drug monitoring Th-cell T-helper lymphocyte TIBC total iron binding capacity TTT total therapy time (for APD and CCPD) TTV total therapy volume (for APD and CCPD) TZD thiazolidinediones (glitazones) U&Es urea and electrolytes UKM urea kinetic modelling URR urea reduction ratio US ultrasound USS ultrasound scan UTI urinary tract infection VLPD very low-protein diet WCC white cell count WG Wegener’s granulomatosis 1

What are the functions of the kidney?

Caroline Ashley

The main functions of the kidneys may be Terms summarised as follows: • Renal capsule: the membranous covering • Regulation of the water and electrolyte con- of the kidney tent of the body by filtration, secretion and • Cortex: the outer layer over the internal reabsorption medulla. It contains blood vessels, glomeruli • Retention of substances vital to the body (which are the kidneys’ ‘filters’) and tubules, such as protein and glucose and is supported by a fibrous matrix • Maintenance of acid/base balance • Hilus: the opening in the middle of the • Excretion of waste products, water soluble concave medial border for nerves and blood toxic substances and drugs vessels to pass into the renal sinus • Endocrine functions – the kidney activates • Renal column: the structures which sup- both erythropoietin and vitamin D. It also port the cortex. They consist of lines of blood produces renin (in the afferent arteriole), vessels and tubules, and a fibrous material which affects various aspects of water and • Renal sinus: the cavity that houses the electrolyte homeostasis. renal pyramids • Calyces: (singular calyx) the recesses in the internal medulla which hold the pyramids. The functional anatomy of the kidney They are used to subdivide the sections of the kidney • Papillae: (singular papilla) the small con- In a normal human adult, each kidney is about ical projections along the wall of the renal 10 cm long, 5.5 cm in width and about 3 cm sinus. They have openings through which thick, weighing 150 g. Together, kidneys weigh urine passes into the calyces about 0.5% of a person’s total body weight. The • Renal pyramids: the conical segments kidneys are ‘bean-shaped’ organs, and have a within the internal medulla. They contain concave side facing inwards (medially). the secreting apparatus and tubules, and are There are three major anatomical demarca- also called malpighian pyramids tions in the kidney: the cortex, the medulla, and • Renal artery: two renal arteries come from the renal pelvis. The cortex receives most of the the aorta, each connecting to a kidney. The blood flow, and is mostly concerned with re- artery divides into five branches, each of absorbing filtered material. The medulla is a which leads to a ball of capillaries. The arter- highly metabolically active area, which serves to ies supply unfiltered blood to the kidneys. concentrate the urine. The pelvis collects urine The left kidney receives about 60% of the for excretion (Figure 1.1). renal bloodflow • Renal vein: the filtered blood returns to the systemic circulation through the renal veins which join into the inferior vena cava

1 2 Chapter 1 • What are the functions of the kidney?

Renal cortex

Fibrous capsule

Minor calyces

Blood vessels entering renal parenchyma Renal medulla (renal pyramid) Renal sinus Renal papilla Major calyces

Renal pelvis Renal column (of Bertin)

Medullary rays Fat in renal sinus

Base of renal pyramid Minor calyces

Ureter

Figure 1.1 The anatomy of the kidney. Right kidney sectioned in several planes.

• Renal pelvis: basically just a funnel, the 2 The proximal convoluted tubule, which re- renal pelvis accepts the urine and channels it absorbs most of the filtered load, including out of the hilus into the ureter nutrients and electrolytes. • Ureter: a narrow tube 40 cm long and 3 The loop of Henle, which, depending on its 4 mm in diameter, passing from the renal length, concentrates urine by increasing the pelvis out of the hilus and down to the blad- osmolality of surrounding tissue and filtrate. der. The ureter carries urine from the kidneys 4 The distal convoluted tubule, which reabsorbs to the bladder by means of peristalsis water and sodium depending on needs. • Renal lobe: each pyramid together with the 5 The collecting system, which collects urine associated overlying cortex forms a renal lobe. for excretion. There are two types of nephrons, those localised to the cortex, and those extending into the medulla. The latter The nephron are characterised by long loops of Henle, and are more metabolically active. The functional unit of the kidney is the nephron (Figure 1.2). Each kidney consists of about one The nephron makes urine by million nephrons. There are five parts of the • Simple filtration – the blood is filtered of its nephron: small molecules and ions 1 The glomerulus, which is the blood kidney • Reabsorption – the required amounts of use- interface, plasma is filtered from capillaries ful materials are reclaimed by selective and into the Bowman’s capsule. passive reabsorption The functional anatomy of the kidney 3

Proximal Distal urine (1% of filtered load). The rate at which convoluted convoluted fluid is filtered by the glomerulus is called the tubule tubule glomerular filtration rate (GFR). The major determinants of GFR are: 1 Renal blood flow and renal perfusion pres- sure 2 The hydrostatic pressure difference between the tubule and the capillaries 3 The surface area available for ultrafiltration. Filtration takes place through the semi- permeable walls of the glomerular capillaries, which are almost impermeable to proteins and large molecules. The filtrate is thus virtually free of protein and has no cellular elements. The Glomerulus glomerular filtrate is formed by squeezing fluid through the glomerular capillary bed. The driv- ing hydrostatic pressure (head of pressure) is controlled by the afferent and efferent art- erioles, and provided by arterial pressure (Figure 1.3). About 20% of renal plasma flow is filtered Loop of Henle each minute (120 mL/min). In order to keep the renal blood flow Cortical collecting and GFR relatively constant, the hydrostatic duct pressure in the glomerulus has to be kept fairly constant. When there is a change in arterial Figure 1.2 The nephron – the functional unit of the blood pressure, there is constriction or dilata- kidney. tion of the afferent and efferent arterioles, the muscular walled vessels leading to and from

• Excretion – surplus or waste molecules and Afferent Glomerulus Efferent ions are left to flow out as urine. arteriole arteriole In 24 hours the human kidneys reclaim 60 mmHg 32 mmHg • ~1300 g of NaCl Glomerular Glomerular hydrostatic colloid oncotic • ~400 g NaHCO3 • ~180 g glucose pressure pressure • almost all of the 180 L of water that entered the tubules.

The glomerulus Bowman’s Renal blood flow is 25% of cardiac output capsule (1200 mL/min). Approximately 99% of the pressure 18 mmHg Bowman’s blood flow goes to the cortex and 1% to capsule the medulla. Renal plasma flow is about 660 mL/min, and 120 mL/min is filtered out of Net filtration pressure = 10 mmHg the blood and into the nephron. Ultimately approximately 1.2 mL of this fluid is excreted as Figure 1.3 Glomerular filtration. 4 Chapter 1 • What are the functions of the kidney? each glomerulus. This process is called auto- Most of the solute reabsorption is active, regulation. with water being freely permeable and therefore moving by osmosis. When the active reabsorb- tion of solute from the tubule occurs, there is a Proximal tubule (Figure 1.4) fall in concentration and hence osmotic activity within the tubule. Water then moves because of The function of the renal tubule is to reabsorb osmotic forces to the area outside the tubule, selectively about 99% of the glomerular filtrate. where the concentration of solutes is higher. The proximal tubule reabsorbs 60% of all solutes, which includes 100% of glucose and amino acids, 90% of bicarbonate and 80–90% of Loop of Henle inorganic phosphate, vitamins and water. Reabsorption is by either active or passive The loop of Henle is the part of the tubule transport. Active transport requires energy to which dips or ‘loops’ from the cortex into the move solute against an electrochemical or a medulla (descending limb) and then returns to concentration gradient. It is the main determin- the cortex (ascending limb). It is this part of the ant of oxygen consumption by the kidney. tubule where urine is concentrated if necessary. Passive transport is where reabsorption occurs This is possible because of the high concentra- down an electrochemical, pressure or concen- tion of solute in the substance or interstitium of tration gradient. the medulla. This high medullary interstitial

Convoluted tubules Efferent arteriole Proximal Distal Glucose Glomerulus Amino acids Cl–

H2O

NH3 H+ K+ Na+ HCO – Bowman’s 3 Uric acid + capsule H Lytes Creatinine –2 HPO4 PCN – Afferent HCO3 Urea arteriole H O Cl– 2 Urea

H2O Filtered through glomerulus Water U Collecting duct Electrolytes r Glucose i Amino acids n Urea Loop of Henle e Uric acid + NH Creatinine NaCl NaCl 4 H2PO4

Figure 1.4 Sites of removal or addition of electrolytes from or into tubular fluid. Other functions of the kidneys 5 concentration of solutes is maintained by the Cells within the hypothalamus called osmo- counter-current multiplier system which gives receptors are sensitive to changes in osmotic the kidney the ability to concentrate urine. The pressure of the blood. If there is low water loop of Henle is the counter-current multiplier intake, there is a rise in the osmotic pressure of and the vasa recta is the counter-current the blood; after excess intake of water, the exchanger. The system works as follows: osmotic pressure of the blood falls. Nerve impulses from the hypothalamus stimulate the • The descending loop of Henle is relatively posterior pituitary to produce ADH when the impermeable to solutes but permeable to osmotic pressure of the blood rises. As a result water so that water moves out by osmosis, water loss in the kidney is reduced because ADH and the fluid in the tubule becomes hyper- is secreted, and water is reabsorbed in the col- tonic. lecting duct. • The thin section of the ascending loop of The result of this ‘counter-current multiplier’ Henle is virtually impermeable to water but system is threefold: permeable to solutes, especially sodium and chloride ions. Thus sodium and chloride ions 1 The high concentration of sodium and chlor- move out down the concentration gradient ide (and urea) in the medullary interstitium and the fluid within the tubule becomes first makes this part of the kidney hyperosmolar. isotonic then hypotonic as more ions leave. 2 Fluid delivered to the distal convoluted Urea, which was absorbed into the medullary tubule is hypotonic. So, as this fluid passes interstitium from the collecting duct, diffuses down through this tubule and the collecting into the ascending limb. This keeps the urea duct, it is exposed to very high osmolar press- within the interstitium of the medulla where ures in the surrounding tissues. If the patient it also has a role in concentrating urine. is dehydrated, the pituitary gland produces • The thick section of the ascending loop and ADH, which makes the collecting ducts early distal tubule are virtually impermeable permeable to water, and water is rapidly reab- to water. Sodium and chloride ions, however, sorbed along the concentration gradient. If are actively transported out of the tubule, not, a dilute urine is excreted. making the tubular fluid very hypotonic. A 3 Extracellular fluid volume depends on the sodium–potassium–chloride (1Na+:1K+:2Cl–) amount of sodium in the body, so it is essen- pump actively extracts these electrolytes from tial that the kidney is capable of conserving the tubular fluid in the thick ascending limb. sodium. If the extracellular volume drops, a complex series of neurohormonal interac- tions lead to the release of aldosterone, Distal tubule and collecting ducts which makes the collecting ducts permeable to sodium, which is absorbed. The final concentration of urine depends upon the amount of antidiuretic hormone (ADH) secreted by the posterior lobe of the pituitary. If Other functions of the kidneys ADH is present, the distal tubule and collecting ducts become permeable to water. As the col- lecting duct passes through the medulla with a Acid/base balance high solute concentration in the interstitium, the water moves out of the lumen of the duct The lungs and kidneys work together to produce and concentrated urine is formed. In the a normal extracellular fluid and arterial pH of absence of ADH the tubule is minimally per- 7.35–7.45 (34–46 nmol/L H+ concentration). meable to water so a large quantity of dilute Carbon dioxide (CO2), when dissolved in the urine is formed. blood is an acid, and is excreted by the lungs. There is a close link between the hypothal- The kidney excretes fixed acid and performs amus of the brain and the posterior pituitary. three functions to achieve this: 6 Chapter 1 • What are the functions of the kidney?

1 The majority of the filtered bicarbonate is • Inotropic myocardial stimulation and sys- reabsorbed (90% in the proximal tubule). The temic arterial constriction. H+, released as the tubular secretion of acid, The opposite occurs when fluid overload occurs. forms carbonic acid with the bicarbonate – (HCO3 ). + – ⇔ ⇔ Aldosterone H + HCO3 H2CO3 H2O + CO2 Aldosterone promotes sodium ion and water The enzyme carbonic anhydrase, found in reabsorption in the distal tubule and collecting the proximal tubular cells, catalyses the reac- duct where Na+ is exchanged for potassium (K+) tion to carbon dioxide (CO ) and water 2 and hydrogen ions by a specific cellular pump. (H O). The CO diffuses into the cell where 2 2 Aldosterone is also released when there is a it again forms carbonic acid in the presence decrease in serum sodium ion concentration. of carbonic anhydrase. The carbonic acid This can occur, for example, when there are ionises to H+ and HCO –. The H+ is then 3 large losses of gastric juice. Gastric juice con- pumped out of the cell back to the lumen of tains significant concentrations of sodium, the tubule by the Na+/H+ pump and the chloride, hydrogen and potassium ions. sodium is returned to the plasma by the Therefore it is impossible to correct the result- Na+/K+ pump. Water is absorbed passively. ing alkalosis and hypokalaemia without first 2 Other buffers include inorganic phosphate replacing the sodium ions using sodium chlor- (HPO –), urate and creatinine ions which are 4 ide 0.9% solutions. excreted in urine as acid when combined with H+ ions secreted in the distal nephron. 3 Ammonia is produced enzymatically from Atrial natruretic peptide glutamine and other amino acids, and is Atrial natruretic peptide (ANP) is released when secreted in the tubules. Ammonia (NH3) combines with secreted H+ ions to form a atrial pressure is increased (e.g. in heart failure + or fluid overload). It promotes loss of sodium non-diffusible ammonium ion (NH4 ) which is excreted in the urine. Ammonia produc- and chloride ions and water, chiefly by increas- tion is increased by a severe metabolic acid- ing GFR. osis to as much as 700 mmol/day. Antidiuretic hormone Antidiuretic hormone (ADH) – also known as Hormones and the kidney vasopressin – increases the water permeability of the distal tubule and collecting duct, thus Renin increasing the concentration of urine. In con- trast, when secretion of ADH is inhibited, it Renin is a vital component of the renin– allows dilute urine to be formed. This occurs angiotensin system, and increases the pro- mainly when plasma sodium concentration duction of angiotensin II which is released falls, such as following drinking large quantities when there is a fall in intravascular volume (e.g. of water. This fall is detected by the osmo- haemorrhage or dehydration). This leads to: receptors in the hypothalamus. • Constriction of the efferent arteriole to main- tain GFR, by increasing the filtration pressure Erythropoietin in the glomerulus • Release of aldosterone from the adrenal Decreased oxygen delivery to the kidney stimu- cortex lates the release of renal erythropoietic factor • Increased release of ADH from the posterior which acts on a liver globulin to produce pituitary erythropoietin. Erythropoietin acts as a mitosis- • Thirst stimulating factor and differentiation hormone Conclusion 7 to specifically stimulate the formation of a steep concentration gradient across the plasma erythrocytes from its committed progenitors in membrane, causing wastes to diffuse out of cells the bone marrow. and into the extracellular fluid. Some of the nitrogen is used to manufacture new nitrogen-containing molecules, but most of Vitamin D it cannot be used for this purpose and must be The kidneys produce the active form of vitamin disposed of by the body. Typically, the first D, namely calcitriol (1,25-dihydroxychole- nitrogen-containing molecule that forms is calciferol). ammonia (NH3), which is very water-soluble, forming NH4OH, a strong base, which must be excreted before it raises the pH of the body Other hormones fluids. The two most common substances used The following hormones are degraded or to get rid of excess nitrogen are urea and uric excreted by the kidneys: acid. Ammonia is converted to urea, which is water-soluble and excreted in a water-based • Parathyroid hormone solution. While the major portion of human • Growth hormone nitrogenous waste is in the form of urea, • Secretin humans typically excrete some uric acid too. • Cholecystokinin In a similar fashion, the kidneys are also • Glucagon responsible for the excretion of many water- • Gastrin soluble drugs. • Insulin. Any of these hormones may increase in circula- tion in the patient with renal dysfunction if Conclusion the kidney is unable to excrete or degrade the hormone. In general, humans can live normally with just one kidney, as they have more functioning Excretory functions renal tissue than is needed for survival. Only when the amount of functioning kidney tissue As cells perform their various metabolic is greatly diminished will chronic renal failure processes, protein and nucleic acids, both of develop. If the glomerular filtration rate has which contain nitrogen, are broken down. fallen very low (end stage renal failure), or if the Other metabolic by-products include water and renal dysfunction leads to severe symptoms, carbon dioxide. Chemical groups such as nitro- then renal replacement therapy is indicated, gen, sulfur and phosphorus must be stripped either as dialysis or renal transplantation. As can from the large molecules to which they were be seen from the diverse functions of the kid- formerly attached, as part of the process to pre- neys, renal failure generates a complex group of pare them for energy conversion. This continu- symptoms, which are discussed in the chapters ous production of metabolic wastes establishes of this book.

2

Laboratory tests and investigations

Elizabeth Lamerton

Most patients admitted to hospital undergo a reference range assumes that 95% of the popu- series of standard or routine blood tests per- lation’s values will be within the 2.5–97.5 per- formed on admission and/or during their stay. centile. Reference ranges do not generally take Some of these tests relate to renal function. individual characteristics such as age, sex, Such tests can be used to detect and diagnose weight or disease state into consideration. kidney disease, kidney damage and kidney func- A list of common biochemical tests used in tion and to monitor response to treatment. the biochemistry laboratory to analyse blood Renal patients will have a wide range of tests samples quantitively for a variety of electrolytes and investigations on top of the standard tests. is shown in Table 2.1. It is important to consider each test not in iso- lation but in the context of other results for each individual patient. In this chapter routine Creatinine tests for adult patients will be discussed. Paediatric patients may have additional tests Creatinine is produced continuously in muscle and investigations; these are discussed in more and is a function of muscle mass. The produc- detail in Chapter 13.1–3 tion is usually constant although the rate of In this chapter we will cover: production declines with age and in low muscle mass or cachectic states. Creatinine is elimin- • Common biochemistry tests ated from the body by the kidneys, predom- • Common haematological tests inantly through glomerular filtration, with an • Urine tests additional 30% eliminated by active tubular • Renal biopsy secretion. The plasma concentration of creat- • Immunological tests inine is therefore linked both to body muscle • Radiological investigations mass and the ability of the kidney to excrete the • Classification or staging of chronic kidney creatinine effectively. As a crude indicator, at disease steady state, a doubling of serum creatinine may • Measurement and estimation of kidney indicate a 50% reduction in glomerular filtra- function. tion rate.1–3 Creatinine clearance is routinely used as a measure of kidney function. Standard tests and investigations for the general adult patient population Urea

Each laboratory has a specific reference range Urea is produced when protein or amino for blood test results. When identifying high acids are broken down in the liver. Protein is or low values for your patients, it is essential sourced from both the diet and from body tis- that you refer to the local laboratory range. The sues. Like creatinine, urea is normally cleared by

9 10 Chapter 2 • Laboratory tests and investigations

Table 2.1 Common biochemical tests

Test Sample Approximate reference range for general population

Creatinine Plasma 50–120 µmol/L Urea Plasma 3.0–6.5 mmol/L Potassium Plasma 3.5–5.0 mmol/L Sodium Plasma 135–145 mmol/L Calcium Serum 2.20–2.55 mmol/L Phosphate Serum 0.8–1.6 mmol/L Magnesium Serum 0.8–1.0 mmol/L Albumin Serum 40 g/L Bicarbonate Plasma 20–30 mmol/L Glucose Serum Fasting 3.3–6.7 mmol/L Non-fasting <10 mmol/L Ferritin 24–300 µg/L males 15–300 µg/L females Total iron-binding capacity 45–70 µmol/L Serum iron 12–30 mmol/L

glomerular filtration and is reabsorbed in the both hypokalaemia and hyperkalaemia having kidney tubules. It is a weakly alkaline substance potentially fatal consequences. The intracellular and osmotic diuretic. potassium acts as a reservoir for the extracellu- The urea concentration may be increased fol- lar potassium. lowing consumption of a high protein diet, Normal potassium intake is 60–100 mmol/ states of increased catabolism due to tissue day and comes from the diet via the intestines, damage, sepsis or starvation and by increased the kidneys (glomeruli and tubular cells) and absorption of amino acids, which may occur through cell membranes. following large gastric haemorrhage where Within the kidney, potassium handling blood is digested. occurs through glomerular filtration, tubular When the glomerular filtration rate is reabsorption and tubular secretion. reduced, this is often reflected more rapidly in Potassium undergoes glomerular filtration at serum urea levels than in the serum creatinine concentrations.1 Factors that affect urea are listed in Table 2.2. Table 2.2 Factors affecting serum urea concentration

Factor Effect Potassium Fluid status The total amount of potassium in the adult Oedema (excess fluid) ↓ urea ↑ body is approximately 3000 mmol, most of Dehydration urea ↓ which (98%) is found within the cells. Although Low protein intake urea they represent a small proportion of total body from the diet ↑ potassium, it is the extracellular potassium ions Concurrent infection urea Gastric blood loss ↑ urea that have the physiological effects on mem- Liver function ↓ liver function ↓ urea brane potential and therefore nerve and muscle Pregnancy ↓ urea function. Changes in extracellular potassium Burns ↓ urea can have an immediate effect irrespective of the Chronic nutritional status Cachectic = ↓ urea intracellular potassium concentration, with Standard tests and investigations for adults 11

Table 2.3 Symptoms of hypokalaemia and hyperkalaemia

Potassium Symptoms (mmol/L)

Hypokalaemia 3.0–3.5 Malaise, muscle fatigue, cramps 2.5–3.0 ECG changes, more prominent malaise, muscle fatigue and cramps <2.5 ECG changes, paralysis of legs or trunk can lead to respiratory arrest Hyperkalaemia >6.5 ECG changes, then ventricular fibrillation and cardiac arrest the same rate as plasma and approximately increase in both sodium reabsorption and 60–80% is reabsorbed by the proximal tubular potassium secretion. In addition to hyper- cells through passive transport. Damage to the kalaemia, aldosterone release can also be stim- renal tubular cells may impair this function and ulated by volume depletion and low plasma result in hypokalaemia. Potassium is secreted sodium osmolality.2,3 in the distal tubules and collecting ducts in exchange for sodium. Hydrogen ions may com- pete in this exchange. Management of hyper- and hypokalaemia in the Some potassium moves into the filtrate in renal dialysis patient the thin descending limb of the loop of Henle; The usual range for random serum potassium however, this is counterbalanced by movement concentrations is 3.5–5.3 mmol/L. Patients of potassium into the medullary collecting with kidney disease are generally less able to ducts. Reabsorption of potassium also occurs in excrete potassium and most patients will have the thick ascending limb of the Loop of Henle. potassium concentrations in the upper limits of Approximately 30% of the filtered potassium the ‘normal range’. The symptoms of hypo- will be reabsorbed here and is linked to sodium kalaemia and hyperkalaemia are summarised in reabsorption. Table 2.3. Approximately 10% of filtered potassium The following checklist of questions should reaches the distal tubule, with 95% of this being be used when dealing with the patient with kid- reabsorbed. The high luminal sodium concen- ney disease: tration, and low luminal chloride concentration stimulates the potassium-chloride co-transporter • Is the potassium result a true value and is it to secrete potassium. In the cortical collecting reliable? Could the blood sampling method duct, potassium is both secreted and reabsorbed, have affected the result? Could the blood and this is the main site of potassium secretion have haemolysed in the sample container? in the kidney. As the plasma potassium concen- • Is this a pre- or immediately post-dialysis tration rises, so does the rate of excretion of reading? Pre-dialysis readings are usually potassium via the kidneys. high and immediate post-dialysis samples are A rise in the potassium level of the low until the patient re-equilibrates (up to 4 extracellular fluid of the adrenal cortex will hours post dialysis). result in the release of aldosterone. In the cor- • Is the patient due to have imminent tical collecting duct, aldosterone promotes the haemodialysis or peritoneal dialysis? High synthesis of Na+/K+ ATPases and their subse- potassium: If the potassium level is not life quent insertion into the basolateral membrane. threatening and haemodialysis is planned This effect on the cell creates an electrical within 12 hours no action may be required. potential favouring the movement of potas- Low potassium: Advise the dialysis team and sium from the cell to the urine. Apical sodium consider amending the dialysis prescription and potassium channel activity is also stimu- to a higher potassium dialysate. lated by aldosterone release, resulting in an • Is there a previous value? Is this acute or 12 Chapter 2 • Laboratory tests and investigations

chronic? Does the patient often have a high giving IV calcium will be seen in 1–5 minutes or low serum potassium? and the duration of action is 1 hour. Repeat if • What are the patient’s other electrolyte necessary after 1 hour. values? Remember always to check mag- For all patients with hyperkalaemia, give nesium in patients with low potassium. therapy to drive potassium into the cells: 10 • Is there a drug cause (e.g. ACE inhibitor, units of soluble insulin in 50 mL of 50% glucose angiotensin receptor blocker, NSAIDs, potas- by IV infusion over 30 minutes. Increasing the sium supplements, amiloride, spironolac- availability of insulin drives the potassium tone)? Can the drug be stopped safely? Will into the cells by enhancing the Na+/K+-ATPase this be enough action? pump in skeletal muscle. Expect a drop in serum potassium of 0.5–1.5 mmol/L and monitor Immediately following haemodialysis a closely for hypoglycaemia. The effect begins patient’s serum potassium levels will be tem- in 15 minutes, peaks at 60 minutes and lasts for porarily lower and levels measured at this time 4–6 hours. An additional dose may be given. will not accurately reflect the actual potassium If necessary prescribe additional emergency ther- level. If necessary potassium should be checked apy to drive potassium into the cells: at least 4 hours after haemodialysis. nebulised 5 mg every 2–3 hours. Take caution • Serum potassium >3.0 mmol/L and with this therapy, as sympathetic activity will be <3.5 mmol/L: Ask the patient if they have a increased causing tachycardia, arrhythmias and potassium-restricted diet. If the answer is yes, fine tremors. temporarily remove the restriction! In addition, sodium bicarbonate may be • Serum potassium <3.0 mmol/L but given, either 1000 mg three times a day orally >2.5 mmol/L: Oral potassium supplements or 500 mL 1.26–1.4% IV (via central or periph- may be indicated. Recheck potassium con- eral line) over 6 hours. Raising the systemic pH centration daily. results in hydrogen ion release from the cells • Serum potassium <2.5 mmol/L: Intravenous and moves potassium into the cells. There is potassium 40 mmol in 500 mL 0.9 NaCl or also a direct effect independent of pH. Effects 5% glucose over 12 hours or, for fluid- begin within 30–60 minutes and last for 6–8 restricted patients, IV KCl 40 mmol in hours. It should be noted that this strategy can 100 mL via central line, over 6 hours. Note cause salt and water overload – exercise caution potassium may also be added to peritoneal in fluid restricted patients. NEVER give bi- dialysis fluid. Recheck potassium level 4 carbonate and calcium together through the hours after the end of the infusion and repeat same line as precipitation will occur. if necessary. A longer-term strategy is to remove potas- sium from the body: Calcium polystyrene sul- For hyperkalaemia K+ >5.3 mmol/L, remem- fonate (Calcium Resonium) 15 g four times a ber that dialysis is the only reliable method of day orally or 30 g rectally. This is an ion removing potassium from the body in dialysis- exchange resin that exchanges calcium for dependent patients. Consider planning dialysis potassium in the gut. This is more useful in at an early stage! Remember to stop any chronic hyperkalaemia than acute hyper- causative medicines or fluids. kalaemia as effects begin in 2–24 hours and last If ECG changes are present (bradycardia, for 24–48 hours after stopping. Therapy must absent P-waves, broad QRS complexes and tall only be given for 48 hours. It can cause severe T-waves) or if K + >7.0, urgent action is required. constipation, therefore always prescribe laxa- Give IV calcium gluconate to stabilise cardiac tives concomitantly and monitor. If the patient muscle and reduce the risk of arrhythmia: has a high serum calcium level, consider using 2.2 mmol = 10 mL of calcium gluconate 10% by sodium polystyrene resonium (Resonium-A) slow IV bolus injection into a large vein. instead. Calcium gluconate is highly irritant and causes severe damage when extravasated. The effect of Urine tests 13

Sodium of calcium, mmol/L] – 0.02 ([albumin concentration, g/L] – 45) mmol/L Sodium is found in the extracellular fluid. There are approximately 3000 mmol of osmotically active sodium in the human body. Normal Haematological tests and investigations sodium intake is approximately 60–150 mmol per day. Net daily losses of sodium are 100 mmol of sodium in urine and 15 mmol in faeces and Some common haematological laboratory tests less than 30 mmol of sodium in sweat. and reference ranges are listed in Table 2.4. 3 Renal blood flow and aldosterone control • Red blood cell count: Erythropoiesis is the sodium balance. Aldosterone exerts its action by production of red blood cells in the bone controlling loss of sodium from the distal renal marrow and is stimulated by a hormone pro- tubule and the colon in exchange for potassium duced in the kidney called erythropoietin. and hydrogen ions.3 Further discussion about erythropoeitin and red blood cell production can be found in Chapter 5 on renal anaemia. Calcium • Haemoglobin: The higher concentration of red blood cells found in men compared with The total amount of calcium in the body women is responsible for the higher haema- depends upon the dietary intake of calcium and globin concentration in men. the calcium lost. The average adult will ingest • Platelets: Another function of the bone 25 mmol of calcium and absorb 6–12 mmol. marrow is the production of platelets. Once Calcium is lost from the body in faeces follow- released into the circulation, platelets have a ing formation of insoluble complexes with relatively short lifespan of 8–12 days. phosphate or fatty acids in the intestine. • White cell count: White blood cells or leuko- Urinary loss of calcium is a function of a com- cytes contain nuclei. There are two main bination of glomerular filtration and tubular types – granular and agranular. The granular function. leukocytes may be identified by staining and Calcium is present in the plasma as both free are subdivided into eosinophils, basophils ionised calcium, which is the physiologically and neutrophils. Agranular leukocytes com- 2 active form, and as protein-bound calcium. The prise T-lymphocytes, B-lymphocytes and amount of calcium present in the body that is monocytes. not bound to albumin and that is therefore physiologically active is known as corrected calcium. Some laboratories measure total plasma cal- cium concentrations. To calculate the concen- Table 2.4 Haematological blood tests tration of physiologically active calcium, the following formula may be used to correct for Test Reference range the percentage of calcium that is bound to albumin:3 Haemoglobin 13.5–18.0 g/dL (males) 12.0–16.0 g/dL (females) White blood cell count If the serum albumin level is <40 mmol/L: (WCC) 3.5–11.0 109/L Corrected calcium = [total concentration Red blood cell count 4.5–6.5 1012/L (males) of calcium, mmol/L] + 0.02 (40 – [albumin (RBC) 4.4–6.0 1012/L (females) concentration, g/L]) mmol/L Platelets 150–400 109/L Prothrombin time (Pt) Measured in seconds If the serum albumin level is >45 mmol/L: Activated prothrombin time (APTT) 28–34 seconds Corrected calcium = [total concentration 14 Chapter 2 • Laboratory tests and investigations

Urine tests Volume of urine produced in 24 hours. A patient with normal kidney function should Urine is relatively easy to investigate in a vari- produce 2–5 L of urine in 24 hours, depending ety of ways and has the advantage usually of on the patient’s hydration status. For example, providing a non-invasive test and investigation. a dehydrated patient will produce significantly Urine can be easily tested using urinalysis dip- less urine. Oliguric patients produce <500 mL in sticks to detect: 24 hours; anuric patients produce <200 mL • Protein in 24 hours; and polyuric patients produce • Microscopic haematuria (traces of blood) >5000 mL in 24 hours. • Sugar The 24-hour urine collection can also be • Infection. qualitatively analysed for protein concentration, cortisol, osmolality, phosphate, potassium and This test is easy to do and convenient and thus sodium. is commonly perfomed at home, in the GP sur- gery, outpatient clinic or at the bedside. Blood and protein should not routinely be Urine microscopy present in urine although this is relatively com- mon in people with kidney disease. Protein or Samples of urine can be examined under the albumin in the urine detected on dipstick tests microscope to detect red blood cells, white may indicate bladder infection or indeed kidney blood cells, crystals and bacteria. The identifica- disease and is suggestive of glomerulonephritis tion of red cell casts in the urine is diagnostic of or vasculitis. Microalbuminuria is diagnosed if glomerulonephritis. the albumin:creatinine ratio is greater than or equal to 2.5 mg/mmol in men or 3.5 mg/mmol in women. Urinary protein. Protein excretion of less than 150 mg per day is Twenty-four-hour urine collection considered to be normal. Approximately 30 mg of this amount will be albumin. Persistent pro- Most patients with kidney disease provide a tein loss of more than 150 mg per day suggests 24-hour urine collection for analysis where renal or systemic disease. possible. The volume of urine collected over a Microalbuminuria is the presence of albumin 24-hour period provides a useful indication of in the urine at quantities above the normal renal function. The collection can be both range of 30 mg/L but below the amount quantitively and qualitatively analysed for: (approximately 300 mg/L) detected by dipstick. As proteinuria increases, the albumin forms a • The actual volume of urine produced in a relatively larger fraction of the total protein loss. strict 24-hour time period Proteinuria is best measured on an early morn- • Urinary creatinine clearance ing spot urine sample to enable calculation of • Urinary protein the protein-creatinine ratio (PCR). Relating the • Calcium urine protein loss to creatinine enables a cor- • Oxalate rection for variations in urine concentration, • Uric acid. and is an important prognostic factor in the When giving a patient instructions on how management of kidney disease. In many cases, to take a 24-hour urine, it is important to make single samples may be used in place of a 24- sure they understand that they should go to the hour urine collection. toilet as usual first thing on the morning of the Unexpected abnormal results should be collection, then collect the urine for the whole repeated at least three times. Proteinuria greater of the day and first thing the next morning. than 3.0 g in 24 hours is considered to be in the That completes the 24-hour collection. nephrotic range. Serum and urine electrophoresis 15

Renal biopsy Routine tests

Routine tests include: Renal biopsy may be performed to diagnose the cause of the kidney disease. Native kidney bi- • Antineutrophil cytoplasmic antibodies opsies are usually carried out in the radiology (ANCA) to look for systemic vasculitis such as department under radiological guidance to Wegener’s granulomatosis or microscopic ensure accuracy. polyangiitis. Those patients with Wegener’s Prior to the biopsy, anticoagulant and granulomatosis exhibit c-ANCA reactivity antiplatelet therapy must be discontinued to again proteinase 3 (PR3), while those with reduce the risk of post biopsy bleeding. Local microscopic polyangitiis have p-ANCA reac- policies should be followed, but common rec- tivity against myeloperoxidase (MPO). ommendations include: • Antiglobular basement membrane antibodies (anti-GBM) as markers of Goodpastures’s dis- • Stop aspirin or clopidogrel for 7 days ease • Avoid NSAIDs for at least 24 hours before and • Double-strand DNA (anti-dsDNA) as a marker after biopsy of systemic lupus erythematosus (SLE). • Stop coumarin anticoagulant therapy for at • Complement factors – C3 and C4 to look for least 2–5 days and replace with heparin if systemic lupus erythematosus, infective continuous anticoagulation is required endocarditis, cryoglobulinaemia • Continue antihypertensive medication and • Myeloperoxidase (MPO) is positive in 80% of ensure blood pressure is adequately con- cases of pauci-immune crescentic glomerular trolled. nephritis and systemic vasculitis In some units, prophylactic desmopresssin • Immunoglobulins – IgG, IgA (DDAVP) is given to control uraemic bleeding, • Bence Jones proteins, light chains, heavy which is often abnormal when urea is greater chains and kappa bands, which can indicate than 18 mmol/L. This is an unlicensed indica- amyloid or myeloma. tion for desmopressin (see Table 2.5). Factor VIII concentrations will be increased two- to fourfold within 1 hour of administration and doses can be repeated every 4 hours if Serum and urine electrophoresis necessary Serum protein electrophoresis is used as a screening tool to detect monoclonal proteins Immunological tests (M-proteins), which can be classified into six general regions, namely albumin, α1, α2, β1 β2 and γ immunoglobulins. The presence of autoimmune antibodies in Albuminuria is usually associated with renal blood or urine detects immunological disease. disease. Microalbuminuria is defined as protein These diseases are discussed in detail in Chapter loss between 3 and 300 mg in 24 hours and is 14. Selected blood tests may indicate specific often seen in patients with diabetes or hyper- disease or diagnoses. tension.

Table 2.5 Unlicensed use of desmopressin in renal biopsy

Agent Dose Onset of action Peak effect Duration

IV desmopressin 0.3–0.4 µg/kg in 50 mL of Less than 60 min 1–4 h 4–8 h 0.9% NaCl over 20 min 16 Chapter 2 • Laboratory tests and investigations

Bence Jones proteins are lights chains stenosis. The renal angiogram is a means of excreted in the urine in patients with myeloma. investigating the anatomy of the renal vascular Bence Jones proteins are not detected using dip- bed. stick urinalysis. Renal angiograms carry an associated risk of acute renal failure caused by the use of X-ray contrast media. This is termed contrast nephro- pathy and will be discussed in Chapter 3. Radiological tests

Renal imaging is an essential investigation in Intravenous excretory urogram renal medicine. Imaging tests are used to invest- igate the size and location of kidneys and look The intravenous excretory urogram (IVU) is a for structural damage or blockages. This is espe- radiological test that uses radiocontrast media to cially useful to detect hydronephrosis suggestive produce a series of radiological images. This test of obstruction – a potentially reversible cause of will identify renal perfusion, structural abnor- acute renal failure. malities and the adequacy of lower urinary tract Imaging techniques include ultrasound, com- voiding. It is generally performed in children or puted tomography (CT), isotope scans and adolescents to ascertain perfusion abnormalit- intravenous pyelograms (IVP). X-ray procedures ies, especially reflux nephropathy, where urine include intravenous urograms, mictuating returns to the kidney and can cause scarring of cystograms, magnetic resonance imaging (MRI) the nephrons and chronic kidney damage. and renal angiograms. Isotope scans such as chromium-labelled ethylenediamine tetra-acetic acid (Cr – EDTA) can assess both overall and Describing and measuring renal individual kidney function within a given per- function son, and are currently the most accurate way of assessing renal function. The concentration of serum creatinine, as dis- cussed earlier, is determined not only by the rate X-ray and ultrasound of renal excretion of creatinine but also by the rate of production, which is in turn dependent The easiest and often first radiological tests are upon muscle mass. Thus serum creatinine may a plain radiograph of the kidneys, ureter and be above the upper limit of normal in patients bladder to look for renal calculi, accompanied with normal kidney function but higher than by a renal and bladder ultrasound. average muscle mass, often observed in young A healthy adult with normal kidneys would men, and similarly the serum creatinine may be expected to have two kidneys, measuring remain within the reference range despite approximately 12 cm in length and of equal marked kidney disease in patients with low size. Small kidneys may indicate either a con- muscle mass, such as slight elderly women. This genital abnormality or long-term (chronic) variation is illustrated in Table 2.6. damage. Large kidneys may indicate the pres- Traditionally biochemistry laboratories have ence of cysts. This can be seen in patients with reported serum creatinine as a measure of kid- polycystic kidney disease. ney function. However, this method is not ideal as it is insufficiently sensitive to detect moder- ate chronic kidney disease, and serum creatinine Renal angiogram concentrations can be affected by other factors as discussed above. Assessing the degree of renal Renal angiograms are performed to visualise the impairment usually involves a measurement of renal artery to confirm a diagnosis of renal glomerular filtration rate (GFR) (see Chapter 10 artery stenosis and, if possible, the cause of the for more detail). GFR is considered to be the best Describing and measuring renal function 17

Table 2.6 Patients with serum creatinine of 130 µmol/L – calculated GFR varies with age, gender, body weight and race Patient Gender Age Weight Race Creatinine GFR (kg) (µmol/L) (mL/min/1.73 m2)

1 Male 25 80 Black 130 100 2 Male 75 80 White 130 48 3 Female 25 50 White 130 45 4 Female 75 50 White 130 25 measure of overall kidney function and forms guidelines.4 In the UK the definition and class- the basis of the five-stage classification of ification shown in Table 2.7 has been adopted chronic kidney disease, which determines by most groups, for example the UK Renal management and referral strategies for patients Association, the UK Royal Colleges of Physicians in whom chronic kidney disease has been and GPs, the British Geriatric Society, the identified. Association of Clinical Biochemists and the Kidney disease is usually identified and National Service Framework 2005 for renal described in terms of acute renal failure (ARF) or medicine.5 chronic kidney disease (CKD). As described earlier, many tests and investigations are used to identify kidney disease and to subsequently Measuring glomerular fitration rate describe the stage of kidney disease. Inter- nationally, chronic kidney disease is routinely Glomerular filtration rate (GFR) can be either classified according to the American National measured or estimated. An accurate GFR may be Kidney Foundation K/DOQI clinical practice measured by injecting a small amount of

Table 2.7 Stages of chronic kidney disease

NFK-K/DOQI Comment: GFR or eGFR using validated Guidelines stage of calculations kidney disease (mL/min/1.73 m2)

Patient is at risk of kidney damage >90 but with risk factors for kidney disease 1 Some evidence of kidney damageb with >90 normal or increased GFR 2 Kidney damage evidentb with some eGFR>60 but <89 reduction in GFR 3 Chronic kidney Moderate reduction in GFR 30-60 disease (CKD)a

4 Severe reduction in GFR 15-30

5 Kidney failure or renal replacement therapy <15 (e.g. dialysis therapy)

aChronic kidney disease is defined as either kidney damage or GFR <60 mL/min/1.73 m2 for >3 months. bKidney damage is defined as pathological abnormalities or markers of damage, including abnormalities in blood or urine tests or imaging studies for example patients with diabetes mellitis, microalbuminuria and normal GFR, patients with cysts in polycystic kidney disease. 18 Chapter 2 • Laboratory tests and investigations radioactive substance and monitoring the rate is important to know which calculation has of travel through the kidney, for example inulin been used to determine GFR and to be aware clearance. This test is rarely done in routine that eGFR often gives a lower number. In clinical practice and is reserved for use in clin- recommending drug dosing it is usually the ical trial settings where a more accurate measure CrCl that has been used not the eGFR in the of GFR may be necessary. summary of product characteristics. As methods of directly measuring creatinine Estimated GFR values are increasingly being clearance are cumbersome, the use of a formula reported directly from clinical biochemistry to estimate renal function from serum creat- laboratories in place of serum creatinine. inine is a practical way to assess kidney func- tion.4 A number of methods are available. The two equations most commonly used in practice Limitations of the Modified Diet in Renal Disease are the Cockcroft and Gault equation and the equation in calculating eGFR Modified Diet in Renal Disease (MDRD) equa- The MDRD formula for estimating GFR should 6,7 tion. Both equations have been validated. not be used in all situations. This is particularly important in situations where the creatinine Cockroft and Gault equation production rate is increased or decreased, the volume of distribution is altered, or the creat- Creatinine clearance (CrCl) = (140 – age in inine excretion rate is modified.6,7 years) weight in kg/serum creatinine in MDRD has not so far been validated in the µ mol/L following patient groups or clinical scenarios: multiplied by 1.04 females or 1.23 for males, • Children where creatinine clearance is expressed in • Acute renal failure mL/min. • Pregnancy • Oedematous states • Muscle-wasting disease states Modified Diet in Renal Disease equation • Amputees –0.999 –0.176 • Malnourished patients eGFR = 170 (Pcr) (age) (0.762 if female) (1.180 if African • Asian patients American) [SUN]–0.170 [Alb]+0.318 • Kidney transplants • People with diabetes. where estimated GFR (eGFR) is expressed in mL/min/1.73 m2, Pcr is plasma creatinine con- centration in mg/dL, age is in years, SUN is References serum urea nitrogen concentration in mg/dL and Alb is serum albumin concentration in g/dL. The equation was developed in patients 1. Davison AM, Cameron JS, Grunfeld JP et al. Oxford with chronic kidney disease; they were predom- Textbook of Nephrology, 2nd edn. Oxford: Oxford inantly white and did not have diabetic kidney Medical Publications, Oxford University Press, disease or a kidney transplant. The equation has 1998. been validated for African Americans but no 2. Mayne Philip D, ed. Clinical Chemistry in Diagnosis other ethnic groups. and Treatment, 6th edn. London: Edward Arnold, The term CrCl from Cockcroft and Gault 1994. 3. Trauls Scott L. Basic Skills in Interpreting Lab Data, used to be interchangeable with the term GFR. 2nd edn. Bethesda, MD: American Society of More recently, with the advent of MDRD to cal- Health Systems Pharmacists, 1996. culate eGFR the waters have been muddied. The 4. National Kidney Foundation. KDOQI Clinical eGFR gives the individual’s renal capacity if Practice Guidelines for Chronic Kidney Disease: their body surface area was 1.73 m2, whereas Executive Summary. New York: National Kidney CrCl is what the kidneys are actually doing. It Foundation, 2002. References 19

5. Department of Health. The National Service 7. Froissart M, Rossert J, Jacquot C et al. Predictive Framework for Renal Services. Part Two: Chronic Kidney performance of the Modification of Diet in Renal Disease, Acute Renal Failure and End of Life Care. Disease and Cockroft-Gault equations for estimat- London: Department of Health, February 2005. ing renal function. J Am Soc Nephrol 2005; 16: 6. Kuan Y, Hossain M, Surman J et al. GFR prediction 763–773. using the MDRD and Cockroft and Gault equa- tions in patients with end-stage renal disease. Nephrol Dial Transplant 2005; 20: 2394–2401

3

Acute renal failure

Caroline Ashley

Acute renal failure (ARF) is defined as the rapid Caution is required when interpreting meas- cessation of renal excretory function within a urements of serum creatinine in ARF for several time frame of hours or days, accompanied by a reasons: rise in serum urea and creatinine, and accumula- • Creatinine production depends on muscle tion of nitrogenous waste products in a patient mass, and though normally constant for an whose renal function was previously normal. It is individual, it can increase in patients with usually, but not always, accompanied by a fall in acute muscle injury. urine output. The condition is potentially • Because of the reciprocal relationship reversible, and in routine clinical practice, mea- between creatinine concentration and GFR, surement of serum creatinine is used to follow small changes in GFR close to the normal the changes in glomerular filtration rate (GFR). range have much less effect on serum creat- inine than small changes when GFR is already significantly reduced. Definition • Changes in serum creatinine concentration lag behind changes in GFR. The serum creat- inine may continue to increase for several There are various working definitions of ARF, days after a marked reduction in the GFR, including: even if the GFR has subsequently started to • an increase in serum creatinine of improve. >50 µmol/L • an increase in serum creatinine of >50% from baseline Incidence • a reduction in calculated creatinine clearance of >50% The incidence of ARF is difficult to state pre- • the need for dialysis. cisely because it depends on the parameters by Confusingly, the patient may be anuric (<50 mL which it is defined. The incidence of severe ARF urine/24 hours), oliguric (<400 mL urine/24 (serum creatinine >500 µmol/L) in the general hours), pass normal volumes of urine or may population is estimated to be approximately even be polyuric. The urine produced may be of 70–140 per million of the population,1 and poor quality, however, with very few waste around half of these will require dialysis. Less products. The diagnosis of ARF is made on severe ARF (serum creatinine ≤177 µmol/L or plasma biochemistry, with an elevated serum an increase of 50% above baseline) occurs in creatinine, urea and possibly potassium. about 210/million/year. One hospital survey Oliguria is usually indicative of failure of both revealed some degree of renal impairment in glomerular and tubular function. In contrast to around 5% of all admissions.2 In intensive care chronic renal failure, there is no early loss of units (ICU), however, the figure is much higher, endocrine function. with at least 15% of admissions having renal

21 22 Chapter 3 • Acute renal failure impairment, of which the cause is sepsis in Causes approximately 50% of cases. The financial implications of renal impairment are consider- able: the cost of a survivor who had renal fail- Conventionally, the causes of ARF are classified ure leaving ICU is 70 times that of a patient by renal anatomy into pre-renal, renal and post- without renal impairment.2 renal causes. This approach is somewhat over- simplified, since many cases of ARF have a mixture of pre, post and renal components. Take, for example, a traumatic injury causing Clinical features rhabdomyolysis and ARF. The injury and associ- ated muscle swelling causes a fall in effective Since ARF involves the acute retention of arteriolar blood volume (EABV) and hence pre- nitrogenous waste products, salt, water, potas- renal impairment. The myoglobin released from sium and acid, the physical signs and symptoms the muscle causes renal vasoconstriction (also encountered include: pre-renal), tubular injury (renal) and tubular obstruction (post renal).3 Nevertheless, since • nausea and vomiting there is no alternative classification in clinical • peripheral oedema use and it is a useful way of considering the kid- • breathlessness ney, the causes of ARF will be described under • pulmonary oedema these headings. • itching • pleural effusion • weakness Pre-renal failure • pericarditis • depression of consciousness Pre-renal ARF is caused by inadequate perfusion • oliguria of essentially normal kidneys, in which the • convulsions. EABV is reduced. It is a normal physiological An episode of ARF usually lasts between 7 and response to hypotension or hypovolaemia, 21 days providing the primary insult is cor- resulting in intense renal conservation of rected in a reasonable time. sodium and water at the expense of a decreased Irreversible ARF usually occurs either in GFR. Renal function usually returns to normal patients with pre-existing renal disease or in rapidly once the underlying cause is corrected. those who experience repeated ischaemic or The kidneys are adept at regulating their nephrotoxic insults. blood supply over a variety of perfusion pres- The mortality rate for ARF is variable. sures, and such autoregulation is highly effect- Patients with non-oliguric ARF have a relatively ive in healthy individuals. This means that low mortality (10–40%), possibly because they quite severe perturbations of blood pressure or have less severe underlying disease or perhaps interference with the usual adaptive responses because they have been treated more promptly of the kidney are required to cause renal dys- or aggressively. A particularly high mortality function in the normal kidney. The operative rate (80–90%) is found in older patients and in word here is normal, since in disease states, for those with serious complications such as pre- example hypertension, this autoregulation may existing cardiovascular or respiratory disease, be disordered or reset, leading to renal dysfunc- severe burns, hepatorenal syndrome, sepsis and tion at blood pressures that would ordinarily be multi-organ failure. quite adequate to maintain renal perfusion. Table 3.1 lists some of the causes of ARF. The traditional signs of sodium and water depletion include tachycardia, hypotension, postural hypotension, reduced skin turgor, reduced ocular tension (sunken eyes), collapsed Causes 23

Table 3.1 Some of the causes of acute renal failure

Hypovolaemia Trauma, burns, surgery, pancreatitis, haemorrhage, gastrointestinal losses, exudative dermatitis, liver failure, nephrotic syndrome, hepatorenal syndrome Loss of peripheral resistance Sepsis, endotoxaemia, shock, general anaesthesia, overuse of antihypertensives, in which the vascular bed is anaphylactic shock, surgery dilated thereby reducing the circulating volume Decreased cardiac output Cardiogenic shock, heart failure, pulmonary embolism, myocardial infarction, cardiac arrhythmias, post-cardiac surgery Renovascular obstruction Atherosclerosis, thrombosis, embolism, dissecting aneurysm Altered renal autoregulation NSAIDs, ACE inhibitors, ciclosporin, tacrolimus peripheral veins and cold extremities. One of injury to renal tubules. This leads to a pro- the physiological responses is a reduction in longed reduction in GFR that sometimes persists renal perfusion, which in turn may lead to for weeks after correction of the initiating intrinsic renal damage with a consequent acute insult. However, the condition is potentially deterioration in renal function. This state may recoverable, provided the initial insult to the be caused by a significant haemorrhage, or by kidneys is removed, and renal perfusion is septicaemia, in which the vascular bed is dilated maintained. thereby reducing the effective circulating vol- ume. It may also be caused by excessive sodium and water loss from the skin, urinary tract or Intrinsic renal failure gastrointestinal tract. Excessive loss through the skin by sweating occurs in hot climates and is Intrinsic renal failure is caused by any factor rare in the UK, but it also occurs after extensive that causes damage either to the kidney itself or burns. Gastrointestinal losses are associated the surrounding vasculature. Table 3.2 lists the with vomiting or diarrhoea. Urinary tract losses numerous mechanisms that can lead to intrin- often result from excessive diuretic therapy but sic ARF. may also occur with the osmotic diuresis caused Renal causes of ARF can be be subdivided by hyperglycaemia and glycosuria in a diabetic into four categories; vascular, glomerular, tubu- patient. lar and interstitial. Infection causes a large proportion of ARF by causing the systemic inflammatory response Vascular syndrome (SIRS). SIRS can be precipitated by a variety of organisms (including bacteria, viruses Blockage of renal blood vessels caused by and fungi) and can lead to multi-organ failure atheroembolic disease or foreign material lead- which has a mortality in excess of 60%.4 The ing to an inflammatory reaction which obliter- mediators of multi-organ failure include haemo- ates the lumen (e.g. cholesterol emboli) can dynamic changes (principally systemic hypo- cause ARF. Occasionally, endothelial damage tension and altered tissue bed perfusion), causes intimal proliferation and luminal oblit- complement activation and cytokine release. eration (e.g. in scleroderma renal crisis or accelerated phase hypertension). The vasculitides cause inflammation and Acute tubular necrosis necrosis in the vessel wall upstream of, or in, Acute tubular necrosis (ATN) comes from the the glomerular tuft (after all the glomerulus is insults that cause pre-renal ARF, but in circum- merely a modified blood vessel). The size of the stances lasting long enough to cause ischaemic vessel involved determines the symptoms and 24 Chapter 3 • Acute renal failure

Table 3.2 Mechanisms that can lead to intrinsic acute renal failure

Acute tubular necrosis General surgery Cardiac surgery Vascular surgery (e.g. repair of abdominal aortic aneurysm, involving cross- clamping of the aorta) Obstetric complications Sepsis Acute heart failure Burns Nephrotoxicity Including aminoglycosides and amphotericin Intravascular coagulation Including hypertension, pre-eclampsia, eclampsia, haemolytic–uraemic syndrome (HUS), thrombotic thrombocytopenic purpura (TTP), scleroderma, disseminated intravascular coagulation (DIC), sepsis Acute tubular necrosis Post-ischaemia Nephrotoxins (drugs, contrast media, organic solvents, herbal medicines, snake venom, mushrooms, heavy metals) Myoglobinaemia Hypercalcaemia Contrast nephropathy A specific form of nephrotoxicity characterised by renal vasoconstriction and avid sodium retention Impaired renal perfusion + ACE inhibitors, NSAIDs, plus atherosclerotic renal vascular disease or drug-induced impairment hypovolaemia of autoregulation Hepatorenal syndrome Reversible intense renal vasoconstriction and sodium retention complicating cirrhosis Poisoning Including paracetamol, often after recovery from liver damage Rhabdomyolysis Following crush injury, drug overdose, status epilepticus Atheroembolism Spontaneous or a complication of angiography, angioplasty or thrombolysis (cholesterol embolism) Raised intra-abdominal Caused by intra-abdominal pressure >25 mmHg (e.g. post-operative abdominal pressure (abdominal exploration, tense ascites) compartment syndrome) Renal embolism Endocarditis, cardiac thrombus Infiltration Including leukaemias, lymphoma, multiple myeloma Urate nephropathy Complication of chemotherapy for acute leukaemia or lymphoma Myeloma Cast nephropathy, light-chain deposition disease, amyloidosis, sepsis and hypercalcaemia can all cause renal damage Hypercalcaemia Sarcoidosis, myeloma Intravenous immunoglobulin Probably results from osmotic damage to proximal tubular cells caused by sucrose in some IV immunoglobulin preparations Renal parenchymal disease Rapidly progressive glomerulonephritis (systemic vasculitis, Goodpasture’s syndrome, systemic lupus erythematosus, Wegener’s granulomatosus) Acute interstitial nephritis Haemolytic uraemic syndrome Cryoglobulinaemia → Causes 25

Table 3.2 (continued)

Renal parenchymal disease Acute allergic tubulo-interstitial nephritis (penicillins, NSAIDs, recreational drugs) (continued) Bacterial endocarditis Infections (Legionnaires’ disease) Granulomas (tuberculosis) Crystals (hyperuricaemia, hypercalcaemia) Malignant hypertension Untreated primary (‘essential’) hypertension or a complication of chronic glomerulonephritis, or scleroderma renal crisis Renal vein thrombosis Complication of malignancy or pre-existing nephritic syndrome Acute pyelonephritis Seldom causes acute renal failure, although may be more likely to do so in the elderly and in those taking NSAIDs Infection in patients with diabetes and partial obstruction (e.g. from papillary necrosis) may cause pyelonephritis and acute renal failure signs and also provides a way of classifying urine output, although the reasons for this have these diseases. not been completely elucidated. Certainly when Microangiopathic haemolytic processes are tubular cells are damaged they slough off the processes in which endothelial damage is the tubular basement membrane into the tubular prime mover, leading to activation of coagu- lumen causing some degree of tubular obstruc- lation, red cell destruction, tubular obliteration tion. In addition, glomerular filtrate is not con- and downstream necrosis. The classical strained within the tubular lumen and leaks examples include pre-eclampsia and the back into the capillaries without change in com- haemolytic uraemic syndrome. position. Finally, and probably most import- antly, renal blood flow is reduced in ATN and blood is diverted towards the medulla away Glomerular from the cortex, bypassing the glomeruli. Renal The glomuruli may be affected by various usu- vasoconstriction is caused by, among other ally immune-mediated insults classified by their things, tubuloglomerular feedback from histological appearance (the glomerulonephri- increased sodium chloride delivery to the tides). These can present either as the nephrotic macula densa, sympathetic stimulation, angio- syndrome (proteinuria >3 g/24 hours, oedema, tensin II, endothelin and thromboxanes. hypoalbuminaemia) with or without renal dys- Metabolic derangements may also lead to function, or as a nephritic illness with nephrotic renal dysfunction, the most common cause of features and/or hypertension and haematuria, which is probably hypercalcaemia, which can often accompanied by renal dysfunction. Drugs cause ARF. Another metabolic problem that are sometimes responsible for inducing can lead to ARF is hypothyroidism. glomerular disease. Interstitial Tubular The interstitium is that part of the kidney that Tubular function may be compromised by is not vascular, glomerular or tubular. As the numerous insults. Tubular cells have adapted to kidney relies on its tightly coordinated structure exist in an ischaemic environment normally, to function, any disruption to this highly inter- but any insult which reduces further the already dependent architecture can result in renal critical supply of metabolites can cause renal failure. An interstitial infiltration with inflam- dysfunction by causing acute tubular necrosis. matory cells including eosinophils is a charac- Tubular damage usually results in a reduction in teristic of many drug-associated cases of ARF. 26 Chapter 3 • Acute renal failure

Acute bacterial pyelonephritis can lead to consequently produce an increase in blood infiltration with inflammatory cells and inter- flow to the glomerulus and the medulla. The stitial scarring and some viral infections are maintenance of blood pressure in a variety of associated with marked interstitial oedema that clinical conditions, such as volume deple- can cause ARF. Autoimmune diseases (e.g. sys- tion, biventricular cardiac failure or hepatic temic lupus erythematosus or mixed connective cirrhosis with ascites, may rely on the release tissue diseases) cause an interstitial infiltrate of vasoconstrictor substances such as that may be irreversible. Very occasionally, the angiotensin II. In these states, inhibition of kidney is infiltrated with cells from lymphoma prostaglandin synthesis may cause un- or leukaemia causing interstitial expansion and opposed renal arteriolar vasoconstriction, ARF. One unusual cause of renal failure is that which again leads to renal hypoperfusion. of the compressed or ‘Page kidney’. In this situ- NSAIDs impair the ability of the renovascu- ation compression of the renal parenchyma by lature to adapt to a fall in perfusion pressure a haematoma (e.g. following a renal biopsy or or to an increase in vasoconstrictor balance. trauma) can cause acute renal dysfunction.5 • Angiotensin-converting enzyme (ACE) Analgesic nephropathy is a special form of inhibitors may also produce a reduction in renal disease in which there is often renal renal function by preventing the angiotensin papillary necrosis and a history of analgesic II-mediated vasoconstriction of the efferent administration. It is now much less common in glomerular arteriole, which contributes to the UK where many of the strongly associated the high-pressure gradient across the drugs have been withdrawn (e.g. phenacetin).6 glomerulus. This problem is important only in patients with renal vascular disease, par- ticularly those with bilateral renal artery Drug-induced acute renal disease stenoses, causing renal perfusion to fall. In Drug-induced renal failure is well recognised, order to maintain the pressure gradient but the frequency with which it occurs with across the glomerulus, the efferent arteriolar particular drugs is unknown. It is, however, resistance must rise. This is predominantly important to be aware of the types of drug that accomplished by angiotensin-induced effer- can induce renal failure because there may be a ent vasoconstriction, as is shown in Figure specific antidote or, if suspected and acted on 3.1. If ACE inhibitors are administered, this early, the failure may be reversible. system is rendered inoperable and there is no Despite a large blood supply, the kidneys are longer any way of maintaining an effective always in a state of incipient hypoxia because of filtration pressure. This leads to a fall in GRF their high metabolic activity, and any condition and the development of ARF. that causes the kidney to be underperfused may • Contrast media, especially the ionic variety be associated with an acute deterioration in (iodinated) used for radiological scans, are renal function. However, such a deterioration also known nephrotoxins, especially in may also be produced by nephrotoxic agents, patients with already compromised renal including drugs. function. There are thought to be two mech- anisms of action behind this toxicity. The • Non-steroidal anti-inflammatory drugs contrast media cause vasospasm which in (NSAIDs) in particular are associated with turn leads to tubular ischaemia and reduced renal damage, and even a short course of an oxygen tension. In addition, oxidative stress NSAID (such as diclofenac) has been associ- causes an increased formation of free radicals ated with ARF, especially in older patients. within the tubules, leading to further The main cause of NSAID-induced renal damage. damage is inhibition of prostaglandin syn- • Iatrogenic factors, including fluid and elec- thesis in the kidney, particularly prosta- trolyte imbalance and drug nephrotoxicity, glandins E2, D2 and I2 (prostacyclin). These can be identified in over 50% of cases of ARF prostaglandins are all potent vasodilators and and also play a large role in many cases Symptoms/signs 27

Afferent arteriole Efferent arteriole Table 3.3 Common causes of obstructive nephropathy

Angiotensin II Bladder outflow Benign prostatic hypertrophy mediates or prostatic carcinoma vasoconstriction of Infiltrative bladder cancer the efferent arteriole causing bilateral vesico- ureteric obstruction Neurogenic bladder Ureteric obstruction Bilateral stone disease Angiotensin II-mediated Crystal deposition (urate, vasoconstriction helps sulfonamides, aciclovir, maintain adequate cisplatin) intraglomerular pressure Pelvic tumours (≥ 35 mmHg) for Papillary necrosis transcapillary Retroperitoneal fibrosis glomerular perfusion (with or without abdominal aortic aneurysm) Figure 3.1 The role of angiotensin II in maintaining Radiation fibrosis adequate intraglomular pressure. Urethral strictures

of community-acquired ARF. Other causes of drug-induced renal failure are discussed in Chapter 11. It has been estimated that up to Symptoms/signs 20% of individuals over the age of 65 are prescribed diuretics, with a lesser number receiving an NSAID; consequently, there is a The initial cause of the ARF is important in large population of elderly patients suscept- determining the symptoms and signs of the ible to renal damage in the event of any presentation. There may be none until late on, insult to the kidney. ARF that requires dia- when signs of fluid overload, oedema and lysis is fortunately rare, with only 50–70 hypertension and hyperkalemia might be pre- patients per million of the population senting features. affected annually, but less severe degrees of Hyperkalaemia may cause cardiac arrest impairment may occur in up to 5% of hos- without warning. Not all patients have the same pital inpatients. susceptibility to the cardiac effects of hyper- kalaemia, but the risk of cardiac arrest can be judged by electrocardiogram (ECG). Pulmonary Post-renal failure (obstruction) oedema is the most serious complication of salt and water overload in ARF, often arising from Post-renal failure or obstructive nephropathy inappropriate administration of intravenous involves obstruction of urinary outflow, leading fluids to oliguric patients. In severe cases, to increased pressure within the renal collecting patients are restless and confused, with sweat- systems and resulting in reduced GFR, reduced ing, cyanosis, tachypnoea, tachycardia and tubular reabsorption of sodium and water, and widespread wheeze or crepitations in the chest. acquired renal tubular acidosis, phosphaturia Further investigation will show arterial hyp- and other abnormalities of tubular function. oxaemia and widespread interstitial shadowing These abnormalities may persist even after relief on chest X-ray. of the cause of the obstruction. Table 3.3 lists the most common causes of obstructive nephropathy. 28 Chapter 3 • Acute renal failure

Distinguishing acute from chronic renal cause of the obstruction should be removed if impairment possible.

One question that must be asked is whether the Fluids renal failure is really acute. Raised serum creat- inine in an acutely unwell patient can be caused In pre-renal failure, urine output and renal func- by acute (ARF), acute-on-chronic, or chronic tion should improve when intravascular volume (CRF) renal failure. These presentations have is restored, thus improving renal perfusion. The different prognoses and may require funda- fluid infused should be blood, colloid or sodium mentally different management. Points to help chloride, and should mimic the nature of the distinguish between them include: fluid lost as closely as possible. Aggressive, early • Comparing any previous measurements of fluid resuscitation is the intervention most serum creatinine with the patient’s current likely to have a positive effect on the course of biochemistry. Pre-existing chronic renal pre-renal ARF and ARF caused by ATN. It must impairment can be excluded if a relatively be guided by regular clinical assessment of the recent previous measurement of renal func- patient’s circulating volume, aided by measure- tion was normal. ments of central venous pressure (CVP) or even • A history of several months’ vague ill-health, pulmonary capillary wedge pressure. nocturia or pruritus, and findings of skin Sodium chloride 0.9% is an appropriate pigmentation, anaemia, long-standing hyper- choice of intravenous fluid, as it replaces both tension or neuropathy suggest a more water and sodium ions in a concentration chronic disease. approximately equal to plasma. Conversely, • Renal ultrasonography to determine size and should water depletion with hypernatraemia echogenicity of the kidneys. It is noteworthy occur, isotonic solutions that are either free of, that renal size is normal in most patients or low in, sodium are available (e.g. dextrose 5% with ARF. or sodium chloride 0.18% with dextrose 4%). • Anaemia is a major feature of CRF, but it may Patients should be observed continuously and occur early in the course of many diseases the infusion stopped ideally when features of that cause ARF. volume depletion have been resolved but before • Bone disease – evidence of long-standing renal volume overload has been induced. bone disease (e.g. radiological evidence of hyperparathyroidism, greatly elevated para- thyroid hormone (PTH) levels) is diagnostic of Fluid balance CRF, but hypocalcaemia and hyperphospha- taemia may occur in both ARF and CRF. Strict fluid balance charts are often ordered in patients with ARF. It is useful to know the fluid input and output, but these charts are notori- ously inaccurate. Over-reliance on them also Management carries the danger that fluid replacement will be adjusted according to the recent output rather Regardless of the cause, the same general treat- than the clinical state of the patient. Positive ment principles apply to all patients who fluid balance is a necessary part of the resusci- develop ARF. These include removing nephro- tation process for patients with effective or true toxic insults (e,g. drugs): in some instances the hypovolaemia. nephrotoxins may need to be removed by dia- However, in extremely overloaded patients, it lysis or adsorption (e.g. after aspirin overdose) may be necessary to restrict fluid intake to the or specific antidotes may be needed in addition urine volume passed in the previous 24 hours + to dialysis (e.g. N-acetylcysteine in paracetamol other losses, for example, insensible losses of overdose). In the case of obstructive ARF, the 500 mL/day. This fluid allowance may be Management 29 increased if the patient exhibits signs of hyper- administration of a fluid bolus to a hypo- ventilation, sweating, fever, wound or drain volaemic patient causes only a transient losses. increase in CVP, whereas a more sustained rise Possibly the most common cause of ARF is is seen in patients approaching euvolaemia. the peripheral vasodilation that occurs in septic shock. In such cases it would be appropriate to Dopamine, loop diuretics and mannitol infuse a colloid as well as sodium chloride as this would help to restore the circulating vol- In ATN, volume repletion does not restore renal ume. It is important to remember, however, that function, and urine output usually remains low not all shocked patients are hypovolaemic and (<30 mL/h). There is no evidence that any treat- some, notably those in cardiogenic shock, could ment improves renal function or accelerates be adversely affected by a fluid challenge. renal recovery. Dopamine has been used at low doses (~2 ng/kg per min) for many years as a reno-protective agent, in the belief that it causes Urine output vasodilatation of the renal vascular bed, restores renal blood flow and thereby improves GFR. Another reason for measuring urine output is to While its use has been controversial, a multi- assess renal function. Anuria and severe oliguria centre randomised double-blinded placebo- are diagnostic of severe ARF, but otherwise urine controlled trial has shown no benefit to using volume is of little help. For example, a GFR of low-dose dopamine infusion in patients with 100 mL/min (6000 mL/h) normally gives a urine renal dysfunction with the systemic inflammat- flow rate of about 60 mL/h because of reabsorp- ory response syndrome.7 The weight of evidence tion of 99% of the filtrate delivered to the renal has swung firmly against the use of low-dose tubules. However, a GFR of 1 mL/min combined dopamine and its routine use in incipient or with complete failure of tubular reabsorption established ARF should be discontinued. (resulting from tubular damage, as in ATN, or The use of a loop diuretic infusion in patients from high doses of drugs that inhibit tubular with non-oliguric ARF has been popular. The reabsorption (e.g. dopamine, furosemide) would theory is that the most metabolically active cells also give a urine output of 60 mL/h. Changes in in the nephron are the first to suffer tubular urine flow are therefore a very poor guide to necrosis in renal underperfusion. Loop diuretics changes in GFR, and must be interpreted along reduce the activity of the Na+/K+/2Cl– pump, with all other available clinical information. which is highly metabolically active, thus Urethral catheters are often placed to enable releasing some metabolic energy towards essen- accurate measurement of urine flow rate. tial subcellular pathways favouring survival in a However, they are the single most important critical incipient tubular cell death. They are source of hospital-acquired Gram-negative sep- also purported to flush out tubular casts. Many sis, so should not be placed without good rea- units give a trial of furosemide (e.g. 500 mg IV son, and should be removed as soon as possible. over 8 hours, or 1 g over 24 hours, if a small dose has produced no effect). Treatment is then continued according to response, although if Fluid challenges the urine output in a euvolaemic patient does not respond to a large dose of furosemide, fur- These are often given in the hope that they will ther therapy should be discontinued in order to stimulate diuresis. It is both illogical and dan- prevent further damage to the tubules.8 gerous to give further fluid to a patient who has Mannitol has been used to promote an already been fully resuscitated. If the patient osmotic diuresis and has been advocated remains oliguric, the extra fluid load may cause by some in cases of incipient ARF and contrast life-threatening pulmonary oedema. The only media-induced nephrotoxicity. However it has rational use of a 500 mL fluid bolus is when not been shown to offer any reno-protective given in combination with CVP measurements; effects in any field of ARF and may cause 30 Chapter 3 • Acute renal failure significant renal impairment itself by causing an volaemia. However, when plasma protein levels osmotic nephrosis and increase tubular work- may be increased as a result of haemoconcen- load by increasing solute delivery.9 tration, crystalloids will probably be more Loop diuretics and dopamine can both effective. In sicker patients with sepsis, endo- increase urine output by reducing tubular re- thelial permeability to macromolecules is often absorption of filtrate, which give the false increased, with the danger that these molecules impression that GFR has improved. The ability will leave the circulation, elevate interstitial to mount a diuresis in response to either drug is oncotic pressure and exacerbate the accumula- probably an indication that the patient may tion of fluid in the extracellular space. have a better prognosis overall. Unless the In the UK, the available colloid solutions patient is significantly volume overloaded, include gelatin preparations, dextran solutions the diuresis caused by these drugs can exacer- and HES solutions. None of the available prep- bate ATN by causing further hypovolaemia. arations are free of potentially serious side- Administration of diuretics to patients with a effects. In particular, dextrans can cause low CVP or to those at risk of hypovolaemia is anaphylactoid reactions and HES solutions may similarly clinically unjustified. disrupt coagulation mechanisms and cause renal impairment. In a trial comparing gelatin with HES 200/0.62 in critically ill patients there Choice of resuscitation fluid was twice the incidence of ARF in the HES- treated group. However, more recent trials Adequate intravascular volume replacement is comparing gelatin solution with the low- essential in the management of ARF. However, molecular-weight, low-substitution HES 130/0.4 controversy continues over the optimal fluid for solution have shown a vastly improved safety correction of hypovolaemia. profile with regard to renal function and coagu- lation with the new HES.10,11 A Cochrane meta-analysis comparing syn- Crystalloids thetic colloids with crystalloids concluded that Crystalloids (e.g. 0.9% sodium chloride, 5% glu- there was no proven clinical benefit for the use cose, Hartmann’s solution) are cheap and safe, of colloids in resuscitation. However, they con- but rapidly distribute between the vascular tinue to be widely used in clinical practice on space and the extracellular space, resulting in the basis of their theoretical advantages. oedema, pleural effusions and ascites if used in large volumes, particularly in the presence of increased endothelial permeability (as in sys- Renal replacement therapy temic inflammatory response syndrome). In cases where the patient has symptomatic meta- Many patients with mild-to-moderate ARF can bolic acidosis, 1.26% sodium bicarbonate solu- be managed on general medical or surgical tion may be infused instead of 0.9% sodium wards, but those with multiple organ failure chloride. The two solutions are both isotonic should be managed on an ICU. Patients with with blood, but the bicarbonate will help cor- ARF requiring renal replacement therapy (RRT) rect the acidosis. (i.e. those with rapidly increasing serum creat- inine, oliguria, especially those with impending or established pulmonary oedema, hyper- Colloids kalaemia and severe metabolic acidosis) should Colloids (e.g. human albumin, dextrans, be managed on either a renal ward or an ICU. hydroxyethyl starch (HES), modified gelatins) There is currently little evidence to guide are used in the hope that they will remain in decisions on when to instigate RRT in ARF. The the vascular space for longer and hence restore options for RRT are usually continuous arterio- microcirculatory flow more efficiently. This venous or veno-venous haemofiltration (CAVH/ theory holds true for cases of simple hypo- CVVH), continuous arterio-venous or veno- Conclusion 31 venous haemodiafiltration (CAVHDF/CVVHDF) A salbutamol nebuliser will lower serum and intermittent haemodialysis. potassium levels transiently by stimulating uptake into cells. Intravenous calcium (10% calcium gluconate, 10 mL over 60 seconds) is given to ‘stabilise’ the cardiac membranes, Additional points but does not alter the serum potassium level. Rapidly acting insulin 10 units (with 50% There are several other practical points to be glucose, 50 mL) given IV over 5–10 minutes, + + considered in the management of patients with stimulates the Na /K -ATPase in muscle and ARF, some of which are especially relevant to liver, driving potassium into cells and reduc- pharmacists. These include: ing the serum concentration by 1–2 mmol/L over 30–60 minutes. This does, however, • Amendment of drug doses: prescriptions render the potassium ‘unavailable’ to a dia- should be closely scrutinised and, where nec- lysis procedure and should therefore be essary, the doses of drugs must be altered so avoided if imminent dialysis is contem- that they are appropriate for the patient’s plated. Cation exchange resins (e.g. calcium level of renal function. Since the level of polystyrene sulfonate (Calcium Resonium) renal function may be in a state of flux in 15 g) orally or rectally 6-hourly may be given someone with ARF, it is important to con- to absorb potassium into the gut lumen. tinue to monitor renal function and re-adjust They require 4 hours to take effect, and lead doses as necessary. to severe constipation if taken without laxa- • In order to allow the already damaged kid- tives, but may be a useful ‘stopgap’ measure. neys to recover, it is also vital to avoid any • Patients with ARF are very prone to develop- further nephrotoxic insults wherever poss- ing stress ulceration, so a proton pump

ible, for example, the use of NSAIDs and inhibitor, H2 blocker or sucralfate should be aminoglycosides. If they must be used, prescribed as per local protocol. ensure that the dose is appropriate, and min- imise the length of treatment. Try to use an alternative, non-nephrotoxic agent wherever Outcome possible. • Maintain adequate nutritional therapy: a good rough guide is a protein intake of Acute renal failure is a life-threatening condi- 0.6 g/kg per day, increasing to 1 g/kg if the tion. In a series of more than 1000 patients patient being dialysed. Calorie intake should requiring dialysis, mortality was 40%.12 Out- be 50–100% above the patient’s resting come depends directly on pathogenesis. If due energy expenditure (25 kcal/kg per day). to obstruction which is successfully relieved, or Consult a specialist renal dietician where ATN where haemodynamic parameters return to possible. normal quickly, then recovery approaches • Monitor the patient’s biochemical parameters 100%. However if due to glomerulonephritis, closely, including daily weight, fluid balance, prognosis depends on severity of damage and U + Es (especially potassium, calcium and may be irrecoverable if too many glomeruli are phosphate), a full blood count, and liver involved. In such cases, the patient will need to function tests. Treat any serious anomalies remain on dialysis permanently. appropriately. • Treat hyperkalaemia if the serum potassium is >6 mmol/L (see also Chapter 2). Severe Conclusion hyperkalaemia with changes in the ECG should be treated as an emergency. Dialysis will correct hyperkalaemia, but it may not be Acute renal failure is common in hospitalised possible to initiate this measure immediately. patients and is most often due to ATN. It is 32 Chapter 3 • Acute renal failure associated with a significant mortality and the patient does not receive any further nephro- morbidity and is expensive to treat. Drugs play toxic compounds during the recovery phase, an important role in the pathogenesis of ARF. and advising on appropriate dose adjustments The pharmacist can play an important role in for drugs given to the patient while undergoing identifying possible pathogens, ensuring that RRT.

C CASE STUDY

Mr VC is a 65-year-old man (68 kg, 175 cm) who presents to casualty with nausea, vomiting and profound diarrhoea. Two weeks ago he presented to his GP with a painful right metatarsal pha- ryngeal joint (due to gout), for which his GP prescribed colchicine 500 µg 4-hourly for 2 days. Unfortunately, the patient became violently sick after four doses and discontinued this medication and went back to the GP who then prescribed: • Indometacin 50 mg three times a day • 150 mg twice daily. The gout pain is now resolving. On admission, Mr VC was pale, lethargic and breathless.

Past medical history: • Hypertension 1 year • Type 2 diabetes 5 years.

Medication: • Bendroflumethiazide 5 mg every morning (for the last six months – increased from 2.5 mg) • Ramipril 2.5 mg every morning (started six months ago) • Gliclazide 40 mg twice daily.

Biochemistry: • Sodium 137 mmol/L (135–150 mmol/L) • Potassium 6.9 mmol/L (3.5–5.2 mmol/L) • Urea 28.5 mmol/L (3.2–6.6 mmol/L) • Creatinine 386 µmol/L (60–110 µmol/L) • Bicarbonate 18 mmol/L (22–31 mmol/L) • Phosphate 1.7 mmol/L (0.9–1.5 mmol/L) • Corrected calcium 2.6 mmol/L (2.2–2.5 mmol/L) • pH 7.26 (7.36–7.44) • Glucose 10.8 mmol/L • 24-hour urine output 600 mL. Mr VC is admitted to hospital under the diabetic team.

→ References 33

C CASE STUDY (continued)

Q1. What patient and pharmaceutical factors may have precipitated acute renal failure in this patient?

Q2. What are the main pharmaceutical problems and how might they be managed? Mr VC is treated for his hyperkalaemia, put on a sliding scale insulin infusion, and rehydrated with 4 L of sodium chloride 0.9%, until he has a central venous pressure (CVP) of 12. After 2 days, his serum creatinine has fallen to 168 µmol/L. However, a further 2 days later, it is noted that he is again failing to pass urine, although he appears to have a palpable bladder, and his serum creatinine has again risen to 272 µmol/L. This suggests urinary retention, so the decision is made to insert a urinary catheter. Following insertion of the catheter, Mr. VC passed 2 L of urine in the next 24 hours, but then spiked a temperature of 38.5°C and became hypotensive. A diagnosis of urinary sepsis was made, and because it was the weekend, and with no other laboratory data available, the new senior house officer (SHO) prescribed gentamicin 475 mg IV daily.

Q3. Comment on the appropriateness of the prescribed antibiotic therapy. What advice would you provide regarding this? Mr VC is given the gentamicin dose of 475 mg IV on the Sunday. On Monday morning, the phar- macist notes that both the drug and the dose are inappropriate for Mr VC, given his degree of renal impairment. The drug is discontinued on the drug chart, but a gentamicin level is taken any- way, and is reported back as 8.6 mg/L, so it is calculated it will take Mr VC several more days to clear that one dose. The prescription is changed to co-amoxiclav IV 600 mg three times a day. His serum creatinine is monitored, and over the course of the next few days, the serial levels are 272, 284, 290, 237, 188, 135 µmol/L. He is switched to oral co-amoxiclav, and his renal function continues to fall. Mr VC is discharged from hospital on day 10, with a serum creatinine of 112 µmol/L.

References 5. Kingdon E, Brunton C, Holt S et al. The re- appearing kidney: an unusual complication of renal biopsy. Nephrol Dial Transplant 1999; 14: 1. Feest TG, Round A, Hamad S. Incidence of severe 1758–1760. acute renal failure in adults: Results of a com- 6. DeBroe M, Elseviers M. Analgesic nephropathy. N munity based study. BMJ 1993; 306, 481–483. Engl J Med 1998; 228: 446. 2. Hou SH, Bushinksy DA, Wish JG et al. Hospital- 7. Australian and New Zealand Intensive Care acquired renal insufficiency: a prospective study. Society Clinical Trials Group. Low dose dopamine Am J Med 1983; 74: 243. in patients with early renal dysfunction: a 3. Holt S, Moore K. Pathogenesis of renal failure in placebo controlled randomised trial. Lancet 2000; rhabdomyolysis: the role of myoglobin. Exp 356: 2139–2143. Nephrol 2000; 8: 72–76. 8. Ho KM, Sheridan DJ. Meta-analysis of frusemide 4. Bone RC. Sepsis and SIRS. Nephrol Dial to prevent or treat acute renal failure. BMJ 2006; Transplant 1994; 9 (Suppl 14); 99–103. 333: 420. 34 Chapter 3 • Acute renal failure

9. Visweswaran P, Massin EK, Dubose TD. Mannitol- Further reading induced acute renal failure. J Am Soc Nephrol 1997; 8: 1028–1033. 10. Langeron O, Doelberg M, Ang ET et al. Voluven, Armitage AJ, Tomson C. Acute renal failure. Medicine a lower substituted hydroxyethyl starch (HES 2003; 31: 43–48. 130/0.4) causes fewer effects on coagulation in Dodds LJ. Drugs in Use, 3rd edn. London: major orthopaedic surgery than HES 200/05. Pharmaceutical Press, 2004. Anesth Analg 2001; 92: 855–862. Kumar P, Clark M. Clinical Medicine, 4th edn. 11. Boldt J, Brenner T, Lehmann A et al. Influence of Edinburgh: WB Saunders, 1998. two different volume replacement regimens on Walker R, Edwards C. Clinical Pharmacy and renal function in elderly patients undergoing Therapeutics, 2nd edn. Edinburgh: Churchill cardiac surgery: comparison of a new starch Livingstone, 1998. preparation with gelatin. Intensive Care Med 2003; 29: 763–769. 12. Liano F, Pascual J. Epidemiology of acute renal failure: a prospective, multicenter, community- based study. Kidney Int 1996; 50: 811–818. 4

Chronic renal failure

Roger Fernandes

This chapter provides a general overview on the The term chronic renal failure (CRF) incidence, causes, consequence and treatments describes a worsening, progressive and irre- of chronic renal failure. The consequences of versible loss of a patient’s kidney function. chronic renal failure, in particular anaemia and Unlike acute renal failure, where a complete renal bone disease, are covered elsewhere in the recovery of renal function is possible, in CRF book and only general principles will be covered the kidneys are permanently damaged, poten- here. The first part of this chapter looks at the tially leading to dialysis or transplantation.1 causes and symptoms experienced in chronic renal failure with a brief discussion on the treat- ment options available for symptom control. The Clinical features second part discusses drug handling in chronic renal failure. The case study at the end highlights some important learning points which should be Chronic renal failure is categorised by severity attempted on completion of the chapter. into grades 1 to 5 (Table 4.1). The severity can be determined by blood tests. This process begins when the glomerular filtration rate (GFR) falls below normal, less than 90 mL/min per Definition 1.73 m2. During progression from acute renal failure In health, the kidney performs a phenomenal (ARF) to CRF the regulatory capacity of the kid- workload: it is perfused with 20% of the heart’s neys attempts to compensate for renal damage output every minute, every day 180 litres of by increasing filtration through the remaining fluid and 1.6 kg of sodium chloride are filtered working nephrons. If the renal function falls out of and then reabsorbed into the circulation, below about 30 mL/min per 1.73 m2, this pro- yet minute changes in dietary salt and fluid cess causes further damage to the kidney and intake are perfectly adjusted for. It primarily generalised wasting. This is often manifested in regulates serum electrolyte levels for sodium, shrinking of kidney size and scarring within all potassium, chloride, calcium, magnesium and parts of the kidney. Patients may often not phosphate amongst others. It stimulates red cell present with any symptoms until over 80% of production from the bone marrow by means of normal kidney function is lost, indicating the erthropoietin and promotes skeletal main- abundant capacity of the organ.1 A proportion tenance by the conversion of vitamin D to of all CRF patients will progress to end stage 25-hydroxycholecalciferol. It is primarily renal failure (ESRF) necessitating renal replace- responsible for the excretion of water-soluble ment therapy in the form of continuous waste products of metabolism. Figure 4.1 shows ambulatory peritoneal dialysis (CAPD), haemo- the glomerulus, the part of the kidney respons- dialysis and/or kidney transplantation (see ible for filtering plasma from capillaries into the Chapter 8). Bowman’s capsule.

35 36 Chapter 4 • Chronic renal failure

Figure 4.1 A normal glomerulus – the sieving unit of the kidney.

Table 4.1 Stages of chronic kidney disease (Renal Association)

Stage GFR (mL/min Description Actions per 1.73 m2

1 90+ Normal kidney function but urine findings Observation, control of blood or structural abnormalities or genetic trait pressure point to kidney disease 2 60–89 Mildly reduced kidney function, and other Observation, control of blood findings (as for stage 1) point to kidney disease pressure and risk factors 3 30–59 Moderately reduced kidney function Observation, control of blood pressure and risk factors 4 15–29 Severely reduced kidney function Planning for end stage renal failure 5 <15 Very severe, or end stage kidney failure Treatment choices Causes 37

Incidence Diagnosis is founded on a detailed medical history, physical examination and a consider- able array of immunological and other sero- Data taken from 22 819 patients from the data- logical tests. Precise diagnosis is important for bases of UK general practitioners in East Kent, identifying and treating potentially reversible West Surrey and Salford showed an estimated causes. It can also assist nephrologists with prevalence of stages 3–5 chronic kidney disease assessing prognosis and planning for replace- 2 of 5.1% in the general population. ment therapy. Kidney biopsies are sometime inconclusive as late in the disease diffuse scar- ring may obscure the primary cause. In these Causes cases or where a biopsy was not performed the diagnosis may be given as unknown. There are three main ways in which kidney Glomerulonephritis is a term used to describe damage can occur: pre-renal, post-renal and a variety of disorders which affect the glomeruli. renal CRF.3 With pre-renal causes, conditions These allow protein and red blood cells that like hypovolaemia, as in major bleeds, or poor normally circulate only in the bloodstream to cardiac function or stenosis of the renal arteries pass into the urine. This clinical picture is often can cause continuous hypoperfusion which may accompanied by hypertension, oedema and ultimately lead to kidney ischaemia and necro- impaired renal function. Clinical presentation sis, resulting in CRF. In post-renal CRF, a dis- ranges from an acute onset which can be cor- ruption of urine flow from the kidneys by rected with treatment, to a chronic insidious bladder obstruction, ureteric stones, retroperi- onset that can progress to established CRF after toneal fibrosis, etc. increases pressure within the many years.4,5 kidneys, eventually damaging nephrons and Polycystic kidney disease (PKD) refers to resulting in CRF. The most common causes of hereditary cystic diseases. Autosomal recessive irreversible damage occur primarily within the polycystic disease is a rare form in children, auto- kidney (renal CRF) and include diabetic nephro- somal dominant polycystic kidney disease pathy, hypertensive nephrosclerosis, vasculitis (ADPKD) presents in adults and the genetic (including lupus and Wegener’s granulomato- defect is present in about 0.1% of the white pop- sis), interstitial nephritis, and polycystic kidney ulation and accounts for about 6% of patients disease.1 Table 4.2 shows data from the 2004 UK presenting for dialysis in the UK. In ADPKD, if Renal Registry. one parent is affected, approximately 50% of

Table 4.2 Percentage primary renal diagnosis by age and gender ratio in England and Wales (Renal Registry 2004)

Diagnosis <65 years >65 years All M:F

Aetiology unc./GN NPa 19.7 29.6 24.6 1.5 Glomerulonephritis 12.9 5.9 9.4 2.4 Pyelonephritis 7.8 7.4 7.6 1.4 Diabetes 20.9 14.9 17.9 1.6 Renal vascular disease 2.4 13.2 7.7 1.6 Hypertension 4.7 5.6 5.1 2.3 Polycystic kidney 9.4 2.7 6.1 1.3 Other 15.7 13.4 14.6 1.4 Not sent 6.6 7.3 6.9 2.4 No. of patients 1992 1942 3934

a GN NP, glomerulonephritis not proven. 38 Chapter 4 • Chronic renal failure their children will develop the disease at some increased gradually whilst monitoring the stage of life.1 In PKD, the kidneys consist of a patient’s blood pressure. The majority of ESRF compact mass of cysts equally distributed patients die from cardiovascular disease. Both through the cortex and medulla causing consid- hypertension and chronic fluid overload can erable enlargement of kidney size. Cysts increase lead to left ventricular hypertrophy (LVH) and in size and eventually rupture, causing infection, systolic and diastolic left ventricular dysfunc- scar tissue formation and hence an overall reduc- tion.1,6 Left ventricular hypertrophy has been tion in functioning nephrons and renal function. shown to be a strong determinant of mortality and morbidity in haemodialysis patients. Hyper- tension also appears to be the major risk factor Symptoms and consequences for the development of atherosclerosis in these patients. Patients experience a higher blood pres- sure and also lose the normal diurnal variation. The symptoms experienced by patients are usu- Treatment aims to correct chronic volume ally a direct sign of uraemia which tends to overload by fluid removal and optimisation of occur as the GFR falls to below 30 mL/min per the patient’s ‘dry weight’. Patients are ‘pre- 1.73 m2.6 The symptoms and signs include scribed’ a maximum amount of fluid to be taken fatigue, electrolyte disturbances, hypertension, per day and dietetic advice is also generally pruritis, ‘restless leg’ syndrome, anorexia, nau- offered on the restriction of sodium in the diet. sea and malnutrition as well as those of anaemia Beta-blockers, calcium antagonists and angio- and of renal bone disease.4 tensin enzyme inhibitors all have evidence of specifically improving left ventricular hyper- 4 Hypertension trophy in the general population. See Chapter 7 for further information. Hypertension occurs in approximately 80–90% of renal patients.1 It can be the cause or result of a decreased renal function. The kidneys Anaemia attempt to autoregulate the pressure of blood entering the glomerular capillaries. Through Anaemia is common in patients with kidney dis- secretion of renin acting on the renin– ease. Healthy kidneys produce erythropoietin angiotensin pathway, the hydrostatic blood which stimulates the bone marrow to produce pressure remains constant, thereby maintaining red blood cells needed to carry oxygen to vital tis- a favourable filtration rate. In renal failure, sues and organs. In CRF, kidney production of falling filtration rates are usually associated with erythropoietin falls, causing a reduction in the sodium and fluid retention and consequent stimulation of red blood cells by the bone mar- hypertension. Conversely, if a patient presents row. Other causes of anaemia in CRF are loss of with hypertension, there is a risk of damage to blood from haemodialysis and low levels of iron the blood vessels in the kidney. The renal artery and folic acid.1 Correcting anaemia can reduce and other smaller vessels become stenosed, lead- lethargy and also decrease left ventricular hyper- ing to impaired blood flow through the organ, trophy, thereby reducing morbidity and mortal- which stimulates renin release. This aggravates ity. the hypertensive state of the patient and accel- Anaemia may begin to develop in the early erates deterioration of renal function. stages of kidney disease, when there is still Tight blood pressure control can often be dif- 20–50% normal kidney function. This partial loss ficult to obtain and may result in patients being of kidney function is often called chronic renal on maximal doses of many antihypertensive insufficiency. Anaemia tends to worsen as kidney agents simultaneously (e.g. a cocktail of a beta- disease progresses. Most patients with end-stage blocker, alpha-blocker, calcium channel blocker, kidney failure have a degree of anaemia present angiotensin-converting enzyme (ACE) inhibitor, and will be on some form of treatment.4 angiotensin II inhibitor and a diuretic). Drugs In patients who present with greater than should, however, be initiated at low doses and 50% loss in renal function and with a low Symptoms and consequences 39 haematocrit level, anaemia is usually caused by With deteriorating renal function, phosphate a reduced erythropoietin level. The European filtration reduces, resulting in high phosphate Best Practice Guidelines 2004 recommend that levels and hence contributing to a low plasma a detailed investigation of anaemia in patients calcium level. Lack of active vitamin D (which with CRF is performed when haemoglobin is necessary for normal absorption and utilisa- levels fall below 11.5 g/dL in adult women, tion of calcium) results in reduced gut calcium 13.5 g/dL in adult men or 12 g/dL in men absorption, which further reduces plasma cal- over 70 years old.7 The investigation will rou- cium levels, resulting in a combined effect of tinely include tests for iron deficiency and hypocalcaemia, hyperphosphataemia and low blood loss in the stool to exclude causes other vitamin D levels, all contributing to the than CRF. increased stimulation of parathyroid hormone The mainstay of treatment is administration secretion.6 This increases calcium release from of a genetically engineered form of erythro- the bones, leading to renal osteodystrophy and poietin (Eprex, NeoRecormon, Aranesp, etc.).7 fractures. The European Best Practice Guidelines 2004 As can be seen from Figure 4.2, a number recommend achieving a target haemoglobin of of abnormalities of calcium and phosphate greater than 11 g/dL within four months of homeostasis occur. Phosphate retention is asso- starting treatment.7 ciated with reciprocal depression of serum cal- Basic erythropoietin supplementation is not cium level.6 sufficient without monitoring of ferritin and per- Reduced calcium absorption occurs as the centage hypochromic red blood cells. Ferritin diseased kidney fails to hydroxylate 25- levels provide an indication of the amount of hydroxycholecalciferol to the more active form iron stored in the body. The current European 1,25-dihydroxycholecalciferol (Figure 4.3). The guidelines aim for a ferritin level no lower than resulting hypocalcaemia stimulates the para- 100 µg/L and a percentage hypochromic red thyroid glands to return the serum calcium blood cells lower than 5%.1 If iron levels are too level to normal. This results in hypersecretion of low, administration of erythropoietin will be of the parathyroid gland and eventually hyper- limited benefit in raising haemoglobin levels and parathyroidism.6 intravenous iron supplementation may be war- The resulting effects are: ranted. Both ‘absolute’ iron deficiency, due to • Osteodystrophy (due to hyperparathy- depletion of iron stores, and ‘functional’ iron roidism) deficiency, due to inadequate mobilisation of • Osteomalacia (due to lack of vitamin D) iron stores to support demand, can be treated • Ectopic calcification. with iron therapy.7 Administration of oral iron is of some benefit in renal patients. Intravenous Management of renal bone disease includes iron supplementation (Venofer and CosmoFer) administration of phosphate binders, such as has been shown to produce better outcomes and calcium carbonate, aluminium hydroxide, seve- response.7 lamer, magnesium carbonate, lanthanum and In addition to erythropoietin and iron, some vitamin D analogues (e.g. calcitriol, alfacalcidol, patients may also need additional vitamin B12 paricalcitol), suppression of parathyroid hor- and folic acid supplementation. See Chapter 5 mone (PTH), parathyroidectomy, control of for further information. aluminium and transplantation, and will be dis- cussed in Chapter 6.

Renal bone disease Oedema Patients with CRF will exhibit some abnormal- ity of electrolytes, potentially causing a varied This can result from sodium and water reten- degree of renal bone disease. tion, and fluid restriction is usually advised to The kidneys aid the homeostatic balance the patient. Careful compliance with fluid between calcium and phosphate in the body. restriction is vital, but often inadequate. An 40 Chapter 4 • Chronic renal failure

CHRONIC RENAL FAILURE

Phosphate retention Low levels of 1α,25-dihydroxycholecalciferol

Hypocalcaemia

Hyperparathyroidism

RENAL BONE DISEASE

Figure 4.2 Abnormalities of calcium and phosphate homeostasis. important consideration is the number of medi- Pruritis cines that are prescribed to fluid-restricted patients as often a considerable amount of fluid Itching is a common uraemic symptom which is needed to take their tablets which forms part can be distressing to the patient, often worse at of their total daily allowance. Oedema can result night and quite difficult to treat. Treatment con- in breathing difficulties if present in the lung sists of control of phosphate and calcium levels, and can also increase load on the heart. hydrating the skin with topical moisturisers and Diuretics can attempt to increase urine output, the use of sedating antihistamines and allowing patients to be able to drink more, but ondansetron.5 this also has corresponding effects on blood electrolyte levels. ‘Restless leg’ syndrome Cholecalciferol Inactive Patients with this condition describe creeping, crawling, prickling sensations in the lower Alfacalcidol limbs, which are often worse at night and Liver Inactive relieved by moving the legs. It is well recognised in patients with iron deficiency.9 Clonazepam, haloperidol, carbamazepine, pramipexole, 25-hydroxycholecalciferol ropinirole or baclofen are of benefit for some Inactive patients.4,5

Kidney 1α-hydroxylase Nausea

Calcitriol Nausea is common in patients with CRF due to (1α,25-dihyroxycholecalciferol) the accumulation of urea and other toxins. Active Routine antiemetics such as metoclopromide, Figure 4.3 Flow diagram for hydroxylation of cholecal- prochloperazine, etc. are effective, with dialysis ciferol to calcitriol. being the best treatment. Influence of chronic renal disease on drug handling 41

Stress ulceration • use of phosphate binders and ion exchange resins. Uraemic patients have an increased risk of The uraemic state can also alter drug hand- gastrointestinal lesions and are therefore pre- ling. This can be affected in three ways: scribed ranitidine or a proton pump inhibitor as prophylaxis. 1 Reduced protein binding 2 Reduced tissue binding 3 Increased volume of distribution. Aspirin, statins

Patients with CRF have an increased risk of Metabolism cardiovascular disease due to diabetes or being on haemodialysis. Therefore they are frequently The main hepatic pathways of drug metabolism prescribed low-dose aspirin and statins to reduce appear to be unaffected in renal impairment. this risk. Statins should be initiated at a low The kidney has some metabolic activity and dose and increased slowly. some of the kidney’s enzymes are thought to have comparable activity to those in the liver.1 This metabolic activity is reduced in CRF and Diet therefore there is reduced hydroxylation of vita- min D. The kidney is also the main site of Patients with chronic renal impairment need metabolism of insulin, so as a patient’s renal regular advice on their diet. This is partly function deteriorates they may require reduced because of their poor appetite due to uraemia doses of exogenous insulin. and general feeling of being unwell and partly through the need to restrict certain foods which contain excess potassium and sodium. Excretion Hyperkalaemia can be life-threatening, and fruit, vegetables and foods generally containing The failure of the excretory function of the kid- water-soluble vitamins are on a restricted list so neys may lead to the accumulation of drugs, 3 patients may require supplements. Renal multi- with consequent toxicity. It is therefore import- vitamins are available and often used in patients ant to decrease this risk by making appropriate who appear deficient in vital vitamins (see alterations either in dose and/or frequency of Chapter 19). dosing of drugs that are renally excreted or preferably ensuring an alternative drug is administered.3 Influence of chronic renal disease on Particular care should be taken with drugs drug handling possessing a narrow therapeutic index. Drugs which have an active metabolite that is excreted renally (e.g. morphine-6-glucuronide) should Absorption also be used with great care or preferably avoided in CRF. Drug absorption can be reduced in patients with chronic renal disease due to:10 Dose adjustments • oedema of the gastrointestinal tract (‘soggy gut syndrome’), When making dose adjustments, important • alteration in gastrointestinal tract transit questions to remember are: time, • alterations in gastrointestinal pH, • What proportion of the drug is normally • vomiting and or diarrhoea, and cleared by the kidney? 42 Chapter 4 • Chronic renal failure

• What proportion is normally cleared through stage renal disease as well as treat the symptoms other routes, such as metabolism? experienced by the patient. Symptoms are • How toxic is the drug? multifactorial and involve a range of body sys- tems, with which patients usually present in These points are discussed in more depth in clinic. Drug therapy is often complex and the Chapter 10. choice and dose is tailored to specific biochem- ical markers and patient compliance. Preserving renal function and delaying pro- Renal replacement therapies gression to dialysis is the primary aim of man- agement and all nephrotoxic agents and factors need to be minimised. Drug handling during The progression to end stage renal disease will CRF alters, and care must be taken in determin- result in patients receiving either a form of renal ing specific effects of drugs in the patient. replacement therapy or palliative treatments. Monitoring is essential and continuous multi- This is covered in detail in Chapter 8. disciplinary input is needed to ensure patients receive the best treatment, care and advice pos- sible. Conclusion Acknowledgement Chronic renal failure is a progressive illness which usually results from a primary renal dis- I wish to acknowledge the help, review and ease or a renal complication of a multi-system additions made to this chapter by Dr Charles disease. The resulting fall in GFR is progressive Soper, Consultant Nephrologist, Mayday and treatment aims to delay progression to end University NHS Trust.

C CASE STUDY

A 72-year-old retired journalist presents with a three-week history of feeling unwell, tired, nause- ated and unable to climb the stairs because his muscles are too weak. He is known to have chronic renal failure (usual creatinine 350 µmol/L), hypertension, ischaemic heart disease and peripheral vascular disease. He last saw his GP about a month ago for hypertension and had been started on irbesartan. In A&E, he is pale, grey and weak. Blood pressure 140/80 with pedal oedema.

Medication: • Calcium carbonate (Calcichew) 2 tabs three times daily • Alfacalcidol 0.25 µg once daily • Irbesartan 75 mg once daily • Amlodipine 5 mg od • Atorvastatin 10 mg at night • Vitamin supplement (Ketovite) 1 tab once daily → References 43

C CASE STUDY (continued)

• Ferrous sulfate 200 mg once daily • Furosemide 500 mg once daily • Epoetin beta (NeoRecormon) 2000 U SC three times a week.

Blood tests: • Urea 49 mmol/L • Creatinine 960 µmol/L • Potassium 7.2 mmol/L • Sodium 139 mmol/L • Bicarbonate 17 mmol/L • Corrected calcium 2.24 mmol/L • Phosphate 2.3 mmol/L • Haemoglobin 9.0 g/dL • WBC 7.0 109/L • Platelets 190 109/L • % hypochromic red cells 6.0.

Q1. What are the metabolic abnormalities?

Q2. What has caused the abnormalities?

Q3. What is the life-threatening complication and how would you treat it?

Q4. What is the rest of his medication for and how much does it cost per year?

References JGG, Warrell DA, eds. Oxford Textbook of Medicine, 3rd edn. Vol. 3. New York: Oxford University Press, 1996; 322–333. 1. Goldsmith D. Management of renal impairment. 7. Sexton J, Vincent M. Managing anaemia in renal Medicine 2003; 31: 52–56. failure. Pharm J 2004; 273: 603–605. 2. O’Donoghue D, Stevens P, Farmer C et al. 8. National Kidney Foundation. Clinical practice Evaluating the prevalence of chronic kidney dis- guidelines and clinical practice recommendations ease in the UK using GP computerised records. for anemia in chronic kidney disease. Am J Kidney Renal Association 2004, Aberdeen, cited in the Dis 2006; 47 (Suppl 3): S1–S145. Renal Registry Report 2004. 9. Kryger MH, Otake K, Foerster J. Low body stores 3. Ashley C. Renal failure – how drugs can damage of iron and restless legs syndrome: a correctable the kidney. Hospital Pharmacist 2004; 11: 48–53. cause of insomnia in adolescents and teenagers. 4. Morlidge C, Richards T. Managing chronic renal Sleep Med 2002; 3: 127–132. disease. Pharm J 2001; 266: 655–657. 10. Renal Disease and Dysfunction. Certificate in Ap- 5. Shaw L. Chronic Renal Failure; The Beginner’s Guide plied Therapeutics, University of Brighton, 2006. to Renal Pharmacy. London: UK Renal Pharmacy http://www.londonpharmacy.nhs.uk/Clinical/ Group, 2001. Applied_Therapeutics_Modules.asp 6. El Nahas AM, Winearls CG. Chronic renal failure 11. Renal Association. UK Renal Registry Report 2004. and its treatment. In: Weatherall DJ, Ledingham http://www.renalreg.com/Front_Frame.htm 44 Chapter 4 • Chronic renal failure

Further reading Renal Association. Adult Section 7: Anaemia in patients with chronic renal failure. Treatment of Adults and Children with Renal Failure: Standards and Feest T. Epidemiology and causes of chronic renal fail- Audit Measures, 3rd edn. London: Royal College of ure. Medicine 2003; 31: 49–52. Physicians: 2002. Available at www.renal.org/ Macdougall IC. How to get the best out of r-HuEPO. Standards/RenalStandards_2002b.pdf Nephrol Dial Transplant 1995; 10 (Suppl 2): 85–91. Morlidge C, Richards T. Managing chronic renal dis- ease. Pharm J 2001; 266: 655–657. 5

Renal anaemia

John Sexton

Anaemia is a common feature of chronic kidney respond to hypoxia by increasing production of disease (CKD), which has a substantial impact a hormone known as erythropoietin, a protein on morbidity and mortality. This chapter whose existence was first speculated about in reviews: 1897 though it was not actually purified until 1975. Once released, erythropoietin then acts • The maintenance of serum haemoglobin in on cells in the bone marrow to stimulate RBC healthy individuals production. During erythropoiesis, erythroid- • The reasons why patients with CKD become progenitor cells incorporate haemoglobin and anaemic develop through immature RBCs called reticulo- • The consequences of anaemia in renal cytes into mature erythrocytes, losing their impairment, if left uncorrected nuclei in the process. The RBCs have a mean • The standards for initiation and treatment of half-life of up to 12 weeks in the circulation, anaemia in renal failure although this is reduced in uraemia, after which • The management of renal anaemia by cor- they are recycled. recting causes, iron supplementation and It is worth commenting here that athletes use of erythropoiesis-stimulating agents seek to train at high altitude because, in addi- (ESAs) tion to developing greater aerobic fitness, the • Iron supplements and ESAs available on the altitude-related hypoxia they experience will UK market result in increased erythropoietin production • The management of patients who fail to and a rise in serum haemoglobin that persists respond to therapy. on return to lower altitudes. The normal adult human body contains a typical 4 g of iron, though this can vary from 2 Maintenance of serum haemoglobin in to 6 g, depending on sex, age and nutritional the healthy individual status. Very little of this iron is free in the circulation: free iron is toxic to many organ systems, but rather it is to be found in three The red blood cells (RBC) or erythrocytes are the main locations: in haemoglobin, in storage and most numerous cells in the blood, and carry an in transport. iron-containing protein called haemoglobin which is responsible for transporting oxygen in the circulation to the peripheral tissues that Iron in haemoglobin need it. The manufacture of erythrocytes, called erythropoiesis, of which typically 2 million are The majority of the iron in the body at any made every second, is controlled by the body in time, about 3 g, is found in haemoglobin within order to maintain serum haemoglobin in a tight the erythrocytes, performing its primary func- range. The peritubular cells in the kidney are tion as a carrier of oxygen from the lungs to the sensitive to falls in serum oxygenation, and they peripheral tissues.

45 46 Chapter 5 • Renal anaemia

Storage iron Much smaller amounts of iron are absorbed from the diet and excreted from the body each There is considerable storage of iron, typically day. Normal iron losses in minor bleeds, men- 1 g, especially in the liver and bone marrow, struation and turnover of the gastrointestinal including that bound within a hollow protein mucosa amount to 1–2 mg, and a similar called ferritin from which it can be released for amount therefore needs to be absorbed from the use. This storage is hard to assess, but measure- gut. The normal diet contains about 10–20 mg ment of serum ferritin, which does not carry or daily of iron, and so it will be seen that only contain significant iron, is considered to pro- 10% or so gets absorbed, though this can rise to vide an indicator which reflects tissue ferritin about 20% during periods of iron demand. and therefore total available iron stores in the Haem iron from red meat is fairly predictably body. Raised serum ferritin may indicate high absorbed, but the absorption of other iron of levels of stored iron but can also be raised for non-haem origin such as in green vegetables other reasons such as inflammation or infec- is much more variable. It is impaired by tea, tion. In these situations, a consideration of the coffee and milk, consumption of which should level of C-reactive protein (CRP) will enable dif- be separated from mealtimes. ferentiation of high ferritin levels. Low serum Absorption is also impaired by phosphate ferritin levels usually indicate insufficient iron binders and antacids, and drugs which raise the stores in the body to supply erythropoiesis; this pH of the gastrointestinal tract such as proton- is known as absolute iron deficiency. pump inhibitors. Iron absorption can be pro- moted, however, by the consumption of fruit drinks containing ascorbic acid with meals. Transport iron Where iron absorption into the body falls below the loss of iron from the body for pro- A small amount of iron, 4–5 mg at any time, is longed periods, absolute iron deficiency will carried around the body bound to the protein eventually develop, followed by functional iron transferrin. Transferrin carries the iron between deficiency, and finally anaemia. This can occur the stores and the erythropoietic cells in the in malnutrition, pregnancy, prolonged minor bone marrow where it is incorporated into new gastrointestinal bleeding, heavy menstruation haemoglobin-containing RBCs and also be- and similar conditions. The iron cycle is shown tween the reticulo-endothelial system, where in Figure 5.1. RBCs are broken down and recycled, back into Anaemia, a serum haemoglobin concentra- stores again. Normally about 20–50% of the tion which has been reduced to a level affect- transferrin is carrying iron at any time, leaving ing tissue oxygenation, is most commonly capacity to quickly mop up any iron entering associated with an inadequate iron intake or the circulation. Transferrin saturation (TSAT, increased losses of blood, such as in menstruat- the ratio of serum iron to total iron-binding ing women or following trauma. Blood trans- capacity), which can be assessed in the lab- fusions may be administered to correct severe oratory, provides a good indicator of iron avail- anaemia, but for most patients, supplementa- ability for erythropoiesis. Like the forklift tion with sufficient iron over time will suffice driver delivering parts to a factory production to restore serum haemoglobin over a period of line, insufficient transferrin iron will limit weeks to months. More rarely in general prac- erythropoiesis; this is known as functional tice, anaemia can present in the absence of an iron deficiency. underlying iron deficiency, due to a failure of Given the typical life of a RBC, about 1% of erythropoiesis for other reasons. This is seen in them are being turned over every day. To do inflammatory conditions such as rheumatoid this, every day, the bone marrow requires 30–40 arthritis, a variety of vitamin deficiencies, some mg of iron for erythropoiesis, and a similar malignancies, or the failure of erythropoietin amount is being recycled back into store again. production in CKD. The development and consequences of anaemia of chronic kidney disease 47

Dietary iron absorption 1–2 mg iron/day Storage iron 1000 mg

ESA Transferrin increases iron 4 mg iron demand

Spleen Iron cycle 120 day life span of Bone marrow erythrocytes releases erythroblast demands 30–40 mg iron/day 30–40 mg iron/day

Erythrocytes 3000 mg iron

Iron losses

Daily GI blood loss Menstruation 1–2 mg iron/day

Figure 5.1 Iron turnover in the body.

The development and consequences iron delivered from its ferritin storage sites of anaemia of chronic kidney disease bound to transferrin. In absolute iron defici- ency, or where the iron needs of erythropoiesis are increased, such as when erythropoietin ana- In addition to the reduced survival of RBCs in logues are administered, transferrin saturation uraemia, the numerous reasons why patients may fall below that necessary to fuel RBC pro- with renal impairment might become anaemic duction, a situation previously referred to as can be divided into two main groups. The first functional iron deficiency. The second group of set of reasons relates to the non-availability of reasons relates to situations in which even if sufficient iron for erythropoiesis: each 1 g/dL sufficient iron were available to supply the rise in serum haemoglobin requires 150 mg of needs of erythropoiesis, this process would be 48 Chapter 5 • Renal anaemia retarded. That is to say, the bone marrow is CKD (see Chapter 4 for stages of CKD), but the unable to utilise adequate iron stores to make prevalence of anaemia in the natural course of adequate numbers of functional RBCs. Of the disease begins to increase once the glomer- course, several or many of these reasons can also ular filtration rate has dropped below exist concurrently with absolute or functional 60 mL/min, as the patients progress through iron deficiency. Failure of the renal peritubular stage 3 CKD. Untreated, most patients with cells to synthesise adequate erythropoietin is an stage 4 or 5 CKD would be expected to have important reason, but there are a variety of some anaemia. Anaemia is more prevalent and other factors that may impair erythropoiesis. severe in diabetic patients, and in those receiv- These reasons are listed in Table 5.1. ing haemodialysis, who have greater iron losses In the light of the above, it should not be due to blood loss in extracorporeal circuits. surprising that anaemia is a common problem The anaemia developed by renal patients has in patients with renal disease, or would be if serious effects on their morbidity and mortality. action were not taken to both prevent and treat Patients will feel tired and there may be a it as it arises. There is some evidence that the deterioration in both physical and cognitive processes that lead to anaemia are at work early functioning, leading to an impairment of qual- in the course of CKD, although reported preva- ity of life. However, it must be remembered that lence varies widely depending on the definition CKD has its major morbidity and mortality of anaemia, any treatments received, co- through cardiovascular ill-health, as blood morbidities and the rapidity of onset of the pressure, lipids, electrolytes and other risk renal disease. factors are often deranged. Anaemia is an inde- Anaemia can manifest itself at any stage of pendent risk factor for the development of left

Table 5.1 Common contributory factors to the development of anaemia in chronic kidney disease

Absolute or functional iron deficiency Dietary inadequacy Anorexia due to nausea of uraemia Dietary restriction to reduce phosphate Poor iron absorption Absorption poor in uraemia Absorption affected by drinking tea with meals, calcium/aluminium phosphate binders, or proton-pump inhibitors Increased blood losses Stress ulceration (minor gastrointestinal losses) of chronic disease, possibly exacerbated by aspirin and NSAIDs Sampling losses for analysis Losses in haemodialysis circuit – EBPG says IV iron usually required in haemodialysis Reduced erythropoiesis Inadequate erythropoietin Reduced renal synthesis in CKD Missed doses of ESAs – hypertensive when dose was due or poor compliance in self-administration Impaired erythropoiesis in presence Inflammation or infection of adequate erythropoietin and iron Uraemia – inadequate dialysis Raised aluminium or parathyroid hormone ACE inhibitors, immunosuppressant drugs Infection/inflammatory conditions

Vitamin B12/folate deficiency

NSAIDs, non-steroidal anti-inflammatory drugs; EBPG, European Best Practice Guidelines; CKD, chronic kidney disease; ESA, erythropoiesis-stimulating agent; ACE, angiotensin-converting enzyme. Assessing the anaemia of chronic kidney disease 49 ventricular hypertrophy. In one study over 12 addition, since until very recently most ESA use months, a fall in haemoglobin of 0.5 g/dL car- had been in patients with severe CKD, and espe- ried three times the risks of a rise in systolic cially those requiring dialysis, outcome meas- blood pressure of 5 mmHg.1 Anaemia also is ures from early prevention and correction of associated with a rise in left ventricular dilata- developing anaemia whilst patients are still rea- tion, heart failure and death. Once coronary sonably healthy are scarce. Several large trials heart disease is established, either as a result of are currently in progress to try to quantify the anaemia or hypertension, diabetes mellitus, dys- cardiovascular benefits of anaemia correction in lipidaemia, uraemia or some other cause, the more representative cohorts of patients, espe- effects of anaemia continue to impair outcomes. cially in such early disease. One other possible By reducing blood oxygen levels in a similar benefit of anaemia correction might be on the manner to cigarette smoking, the existence of progression of the CKD itself. anaemia will not only mean that the heart will In short, more work is needed to quantify: have to work harder to supply the peripheral 1 the overall (especially cardiovascular) risks oxygen demands of the body, but also that the and benefits of prevention and correction of blood reaching it through the coronary arteries renal anaemia, and will contain less oxygen. Anginal attacks will 2 the most appropriate target haemoglobin therefore be more likely, and it has been shown once treatment is initiated that will optimise that the outcomes after myocardial infarction the benefits against risk and costs of anaemia are worse in the presence of anaemia. correction. There is a clear consensus from pooling the trial data of ESAs that the correction of anaemia ‘provides important clinical and quality-of-life benefits whilst substantially reducing hospital- Assessing the anaemia of chronic 1 isations and transfusions’. The avoidance of kidney disease transfusions in populations who may be the future recipients of renal transplants is espe- cially important to avoid pre-sensitisation to The most detailed guidelines on the manage- foreign antigens which might increase the ment of renal anaemia are those produced by chances of graft rejection. Unfortunately, the the joint working group of the European Renal longer term benefits of anaemia correction are Association and the European Dialysis and less clear-cut, especially in the area of cardio- Transplant Association in 2004, and known as vascular morbidity and mortality, and what the Revised European Best Practice Guidelines information exists is conflicting. Older pharma- for the management of patients with chronic cists will remember a similar situation that renal failure (EBPG).2 The UK Renal Association formerly existed with antihypertensive medica- expanded its own guidance in 2006 and pub- tion. Every antihypertensive on the market, to lished Chronic Kidney Disease in Adults: UK obtain a product licence, has been through a Guidelines for Identification, Management and clinical trials process that shows that it is effect- Referral, which includes guidance on renal ive in lowering blood pressure with an accept- anaemia.1 Finally, the UK National Institute for able adverse event profile. Until recently, Health and Clinical Excellence (NICE) published however, few had been tested in trials powered minimum standards for the National Health to show that they produced the intended out- Service in England and Wales in September comes of a reduction in cardiovascular morbid- 2006, the guidelines ‘Anaemia management in ity and mortality.1–3 people with chronic kidney disease’ being avail- So it is with anaemia correction in CKD. It able via the NICE website.3 Similar guidance has should not be surprising that in a group of been published for the United States by the patients with so many cardiovascular risk fac- National Kidney Foundation Dialysis Outcomes tors the correction of just one of these fails to Quality Initiative (NKF K/DOQI). There are make much impact on overall mortality. In slight differences between the different guide- 50 Chapter 5 • Renal anaemia lines in their definitions of anaemia, thresholds ered if the patient has CKD (stage not specified) for treatment, target haemoglobin levels and and a haemoglobin of 11 g/dL or less.3 Unlike other standards but since they all draw on the the other guidance NICE has not restricted care same evidence base a certain commonality is to any particular degree of CKD, though refer- seen and that shared vision is more important ence is made to CKD stage 3 when considering than small differences in approach reflecting the assessment of anaemia. the different needs and views of their intended The assessment of renal anaemia is clearly users. documented in the EBPG but includes an assess- Healthy adults have serum haemoglobin con- ment of: centrations which usually fall in the range of • the nature and severity of the CKD 12–15 g/dL for women and 14–18 g/dL for men, • the actual haemoglobin being currently though different laboratories cite different refer- achieved ence ranges. The EBPG state that the investiga- • iron stores as reflected by serum ferritin (to tion of anaemia in CKD should begin when exclude absolute iron deficiency) haemoglobin concentrations fall below: • available iron as determined by transferrin • 11.5 g/dL in adult women saturation or % hypochromic RBCs (to • 13.5 g/dL in adult men exclude functional iron deficiency) • <12 g/dL in adult men over 70 years of age.2 • other causes/contributing factors towards renal anaemia (see Table 5.1) These guidelines state that the actual degree of • other medical reasons why the patient might CKD is irrelevant. As was discussed earlier, be anaemic.2 anaemia is more common as CKD advances but anecdotal evidence suggests that most renal cen- Once haemoglobin has fallen to 11 g/dL or tres formerly felt a need to set arbitrary serum below, and contributing factors have been dealt creatinine concentrations for either referral to with as far as possible, then all the guidelines anaemia clinics or the prescription of ESAs. In agree that therapy with ESAs and/or iron should the absence of NICE guidance and funding be initiated, as appropriate. streams, limits have often been set to direct treatment to patients for whom the cost-benefit base is clear, or for whom resources can be made Target haemoglobin levels once treatment is available. In many UK centres, resources to treat initiated patients other than those who have started dial- ysis have only become available in the last few In a similar manner to their selection of patients years and the EBPG guidance merely highlights for assessment, the EBPG stress that treatment how artificial this practice was. availability and targets for haemoglobin to be Both Renal Association and NICE guidance achieved should apply to all patients, though take a slightly different approach.1,3 The Renal this is not always the case in current UK prac- Association guidance in 2006 lists anaemia as tice. So, the EBPG consider equally: something to be checked when CKD reaches • Non-dialysing patients with CKD in stages stage 3 (creatinine clearance <60 mL/min) and 1–5 recommends treatment with ESAs for those • Patients with CKD stage 5 receiving patients with a haemoglobin of <11 g/dL. (Older haemodialysis or peritoneal dialysis Renal Association standards had previously only • Transplant patients with chronic renal insuf- set a target haemoglobin of >10 g/dL in 2002.4) ficiency (undefined) and anaemia.2 This guidance no longer conflicts with the EBPG guidance, which itself recommends ESA therapy The target in the EBPG is to raise haemo- when haemoglobin falls to 11 g/dL and all other globin and maintain it at above 11 g/dL, this causes of anaemia have been excluded or cor- target to be reached within four months after rected. The draft NICE guidance simply suggests treatment initiation. The majority of patients that management of anaemia should be consid- should be able to achieve this, but the upper Iron and erythropoiesis-stimulating agents 51

Table 5.2 Targets for iron stores during therapy for renal anaemia

Target EBPG 2004 EBPG 2004 NICE 2006 Guidance population target

Serum ferritin (to avoid absolute >100 µg/L 200–500 µg/L 100–500 µg/L (non-HD) iron deficiency) (and avoid >800 µg/L) 200–500 µg/L (HD) Transferrin saturation (to avoid >20% 30–40% >20% (unless ferritin functional iron deficiency) >800 µg/L) % Hypochromic RBCs (where available) <10% <2.5% <6% (unless ferritin – (better measure of functional iron >800 µg/L) deficiency)

EBPG, European Best Practice Guidelines; NICE, National Institute for Health and Clinical Excellence; HD, haemodialysis; RBCs, red blood cells. limit to be achieved is still unclear. The guide- be avoided. Iron stores may be adequate ini- lines say that pre-dialysis haemoglobin levels of tially, but will fall rapidly once erythropoiesis is higher than 14 g/dL should be avoided in stimulated with ESAs. The guidelines all give patients on haemodialysis, as the blood further standards to ensure that functional and absolute concentrates during haemodialysis, and that iron deficiency does not impair treatment. Of until more is known, patients with diabetes or course, if functional iron deficiency has been heart disease should be restricted to a maximum the cause of the renal anaemia rather than a of 12 g/dL. If angina is a problem or if there is deficiency in erythropoietin, simply replacing a hypoxia due to concurrent airways disease it iron may be sufficient to generate a rise in may be necessary to aim higher. Some younger, serum haemoglobin. Oral iron supplements fitter patients, especially those with families such as ferrous sulfate tablets may be sufficient to raise or work to go to may feel better for many patients to maintain iron stores. with haemoglobins towards 14 g/dL, but the However, in haemodialysis patients where iron evidence base is unclear and there is a law losses are higher, where initiation of ESAs is of diminishing returns in relation to ESA ther- creating an increased demand for available iron, apy – it takes increasingly high doses and there- or where the patient is starting from a position fore cost to raise haemoglobin above 12 g/dL. of depleted stores, intravenous iron therapy The Renal Association guidelines simply recom- may be required. mend maintaining haemoglobin in the range The targets for adequate iron stores for ery- 11–12 g/dL and the NICE guidance a similar thropoiesis are shown in Table 5.2. It will be 10.5–12.5 g/dL, with therapeutic changes made seen that the EBPG gives population targets as and action taken if it falls outside the range well; these will be higher to make sure that the 11–12 g/dL.1,3 majority of patients in the population reach the minimum individual targets.

Iron store targets Iron and erythropoiesis-stimulating ESAs will not work effectively if functional iron agents available for the correction of stores are low, as once transferrin cannot deliver renal anaemia adequate iron to the bone marrow, initially the RBCs will become hypochromic and then erythropoiesis will cease. In addition, since Once a patient’s haemoglobin has fallen below functional iron stores are replenished from 11 g/dL, they will need treatment for their stored iron, absolute iron deficiency must also anaemia. Twenty years ago, the only treatments 52 Chapter 5 • Renal anaemia available were iron dextran infusions, and, once posable injector that may facilitate patient self- these were ineffective, blood transfusions. Many administration. Patients report more favourably renal patients became transfusion-dependent to on issues such as the ‘sharpness’ of the needle maintain haemoglobin. Apart from the infection and ease of injection with particular products. risks and fluid load this carried, the success of ESAs are temperature-sensitive and need to be future transplants is impaired if patients are stored in a refrigerator and the room temperat- repeatedly exposed to foreign antigens. In addi- ure period which the product is considered safe tion, each unit of blood contains up to 200 mg to use within varies between products. of iron. Over time, multiple transfusions in pa- tients without major blood (and iron) losses leads to iron overload and the need to treat patients Route of administration with chelating agents such as desferrioxamine. However, the increasing availability since the For patients receiving haemodialysis, intra- early 1990s of both synthetic recombinant venous administration during dialysis would human erythropoietin and safer intravenous iron avoid the need to give the patient unnecessary preparations has revolutionised therapy to the injections. However, the shorter half-life of extent that renal anaemia is now a manageable epoetin means that the product is less efficient condition in most patients. The recombinant when given intravenously compared with sub- erythropoietins available in the UK are: cutaneous administration – dose increases of 25% or more have been required to maintain • Epoetin alfa (marketed in Europe as Eprex by haemoglobin levels in some studies. This can Janssen-Cilag, now Ortho-Biotech) add considerable cost across a population, and • Epoetin beta (marketed in Europe as many units have traditionally had to administer NeoRecormon by Boehringer Ingelheim, now epoetin subcutaneously even though vascular part of Roche) access is available during the haemodialysis ses- • Darbepoetin (marketed by Amgen as sion. One advantage of Aranesp is that this loss Aranesp). of efficiency is not seen, and thus intravenous Much work has been done to compare dar- administration is possible without incurring bepoetin with epoetin, especially epoetin alfa, extra costs. For all other patients, subcutaneous and a large body of work now exists comparing administration is the only route practical. With the two drugs in different situations. Most the removal of the Eprex licence for subcutane- sources would conclude that the erythropoiesis ous administration in 2002, this product essen- stimulating agents epoetin and darbepoetin are tially was no longer an option for peritoneal broadly similar in terms of ability to achieve dialysis or non-dialysing patients. The sub- target haemoglobin levels, and adverse-event cutaneous route was reinstated in late 2006, but profiles. This should be remembered when con- the summary of product characteristics (SPC) sidering the differences between products, some now says that the intravenous route is prefer- of which are listed below.5 able and should be used wherever possible.

Presentations Frequency of administration

All ESAs are available in a selection of pre-filled Epoetin was originally administered three times syringes providing different strengths to enable a week, with some early studies suggesting that either nurse or patient/carer administration. In attempting to reduce this increased the required addition, Roche market NeoRecormon as a weekly dose and costs. NeoRecormon is now multiple-use vial for ward or satellite unit use, licensed for subcutaneous use in stable patients and cartridges for use in a pen-type admin- at once-weekly dosing, with a possible use as istration device. Amgen supply a smaller range little as fortnightly. Aranesp, because of its of strengths of Aranesp in an automatic dis- longer half-life, can be initiated weekly and Initiating iron and erythropoiesis-stimulating agents therapy 53 reduced to as little as once a month in stable, infusions of up to 20 mg/kg, the so-called total non-haemodialysis patients. dose infusion. In patients who are not receiving haemodialysis, this removes the need to have to repeatedly attend a clinic or hospital, although administration may take several hours. This Iron preparations practice was also formerly commonly practised with Venofer but the off-label (outside the prod- Oral iron preparations such as ferrous sulfate are uct licence) nature of this method of adminis- cheap, readily available, and do increase iron tration has reduced its popularity. The SPCs for availability from the gastrointestinal tract. both iron preparations require administration to Where an absolute iron deficiency has not yet take place where facilities for cardiopulmonary developed, or is not severe, oral iron may be suf- resuscitation are available, and in the case of ficient to restore absolute and functional iron iron dextran, with a doctor in attendance. status. However, once iron deficiency has Venofer remains the dominant product in become established, or continuing losses are ele- the UK; many sources and the EBPG consider it vated, this may not be the case: the EBPG to be safer, faster working, and the product of observe that most haemodialysing patients will choice.2 However, CosmoFer use is considerable require intravenous iron. Oral iron can, how- and the licensed availability of total dose infu- ever, irritate the gastric mucosa and lead to nau- sions offers advantages in some situations. sea, constipation or diarrhoea. Unfortunately, whilst taking iron tablets with food reduces some of these side-effects, it also means that if Initiating iron and erythropoiesis- the patient is taking phosphate binders, which stimulating agents therapy are also taken with food, this will impair iron absorption. In the UK, two main intravenous iron prep- Practice does vary between units, and the guide- arations are available. Although a preparation of lines take slightly differing approaches. The intravenous iron dextran was used formerly, this major decisions to be made in a CKD patient product had a poor reputation for anaphylaxis whose haemoglobin has dropped below 11 g/dL and was replaced by an iron sucrose product. are whether to start iron, ESAs or both. The Iron sucrose, also referred to by other names forthcoming NICE guidance contains helpful such as iron saccharate, is supplied in ampoules algorithms when initiating ESAs and iron ther- containing 100 mg of ferric iron as Venofer by apy.3 In patients with functional iron deficiency Syner-Med. Iron sucrose can be easily adminis- (TSAT <20% or hypochromic cells >6%), then tered as a slow intravenous injection of up to up to 1 g of ferric iron should be administered 200 mg, or slowly infused in a small volume of over a few weeks, unless serum ferritin is high, 0.9% sodium chloride. On first use, a test dose typically greater than 800 µg/L. ESAs can be of 25 mg over 15 minutes is required by the initiated at the same time, or after a month to product licence, and whilst this is good practice see if the iron replacement has had any effect it is not the case in some other countries or on the serum haemoglobin. The SPCs for dar- required in the EBPG. Several doses may be bepoetin and epoetin can be consulted for pre- required to restore iron stores, followed by cise initiating dose regimens, but epoetin is maintenance doses at intervals of weekly to generally initiated twice weekly in the UK in monthly depending on the patient’s iron losses. non-dialysing populations, and three times a More recently, a lower molecular weight iron week otherwise. Darbepoetin is typically initi- dextran than that used previously has been ated weekly before reducing the frequency of licensed as CosmoFer by Vitaline. As with iron administration. Checking blood pressure and sucrose, iron dextran can administered by intra- haemoglobin fortnightly to monthly, haemo- venous injection or infusion. One advantage of globin is typically allowed to rise at about 1–2 CosmoFer is that its product licence allows large g/L per month, increasing and reducing ESA 54 Chapter 5 • Renal anaemia dose to achieve this until the target haemo- and seems to be related to a change in the globin is achieved. formulation or manufacturing process. It cannot be stressed too strongly that ade- Interesting points are: quate iron stores are required if ESA therapy is • The US epoetin alfa made by Amgen was not to be efficient, and any other contributing associated with the same rise in PRCA cases factors to renal anaemia (Table 5.1) need the as Eprex optimum control possible. ESAs are generally • Intravenous use is associated with a much well tolerated, their major dose-limiting lower incidence of PRCA problems being cost and the effects on blood • Use of Eprex in cancer patients did not lead pressure. to a rise in PRCA, possibly because of the shorter durations of therapy that are typical in malignant disease, or possibly there is Failure to respond to therapy some other factor at play in CKD • Ortho Biotech postulate a possible reaction between the stabiliser polysorbate 80 and the Some patients will fail to respond to ESA ther- formerly uncoated rubber stoppers used until apy, for a variety of reasons, mostly already 2003.6 included in Table 5.1 as a contributory cause of the renal anaemia in the first place. The most Ortho Biotech has now reformulated Eprex common reasons for inadequate response are and had produced sufficient data to allow the considered to be either inadequate functional or recent reinstatement of the subcutaneous route, absolute iron stores, or inflammatory disorders. albeit with the proviso that the intravenous However, in self-administering patients non- route is preferable. Pharmacists should remain compliance must be excluded. Even in those vigilant for signs of PRCA in a patient receiving patients who receive ESA therapy from their any ESA therapy. dialysis nurse or community nursing teams, patients may miss doses for a variety of reasons (hypertension, non-availability of the product Summary at the required time, poor communication, etc.). There is a whole chapter in the EBPG on the response to a patient not responding to darbe- In the last 10 years the incidence of renal poetin at >100 µg weekly or epoetin at >20 000 anaemia has declined sharply, due to the devel- units weekly.2 More worryingly is the failure to opment of ESAs and safer parenteral iron ther- respond to ESAs due to the development of anti- apy. The challenge of the future is to develop bodies to ESAs. The most severe manifestation systems that ensure funding streams are in place of this is known as pure red cell aplasia (PRCA), so that all patients requiring ESAs receive them, a severe anaemia where haemoglobin, RBCs and and that units purchase and use ESAs in the reticulocyte counts fall steadily in the presence most efficient way. Newer ESAs are on the verge of normal white cell and platelet counts. No ESA of licensing, and it seems likely that these will can be administered to a patient with PRCA, work their way into clinical practice. Uptake and lifelong transfusions will be required. PRCA will be slower than previously, due to the can occur spontaneously but is most associated increasing commitment of NHS Trusts to exclu- with subcutaneous epoetin therapy for more sive purchasing arrangements, and fears about than four weeks. the comparatively lower safety data of newer A rise in the incidence of PRCA was noted products, especially in the light of the PRCA since 1998, in particular with epoetin alfa experience. However, patients with CKD need (Eprex). As a result, the licence to administer no longer fear the debilitating effects of renal Eprex subcutaneously for renal patients was anaemia and the NHS has delivered effective revoked in most European countries. The cause correction of haemoglobin levels in the major- of PRCA with Eprex is probably multifactoral ity of patients with CKD. References 55

C CASE STUDY

Mrs AP is a 56-year-old woman with chronic kidney disease secondary to type 2 diabetes mellitus, which has been worsening over several years. She smokes 20 cigarettes a day, drinks 4 units weekly, is tired and breathless, and blood tests are performed.

Blood results: • Potassium 5.2 mmol/L (3.5–5.5 mmol/L) • Creatinine 281 µmol/L (<130 µmol/L) • Phosphate 2.21 mmol/L (0.7–1.4 mmol/L) • Calcium 2.05 mmol/L (2.2–2.6 mmol/L) • HbA1c 9.2% • Haemoglobin 10.1 g/dL (12–15 g/dL) • Blood pressure 165/95 mmHg.

Medication: • Atenolol 50 mg every morning • Amlodipine 5 mg every morning • Isosorbide mononitrate SR 60 mg every morning • Glyceryl trinitrate (GTN) spray when required • Sodium bicarbonate 1 g three time daily • Aluminium hydroxide (Alu-Caps) 2 three times daily • Sevelamer 2 three times daily • Alfacalcidol 1µg twice weekly • Co-codamol 30/500 four times daily • Diclofenac 50 mg twice daily.

Q1. The Senior House Officer (SHO) asks if Mrs AP could be started on ESA therapy. Give three reasons why you might advise against this at this point.

Q2. If ESA therapy is to be initiated, what are the choices in terms of initial dose and monitor- ing?

Q3. What factors might contribute to reduced efficiency of ESA therapy in Mrs AP?

Q4. What factors affect the choice of ESA for Mrs AP?

References Practitioners. Chronic Kidney Disease in Adults: UK Guidelines for Identification, Management and Referral. London: Royal College of Physicians, 1. Joint Specialty Committee on Renal Medicine of 2006. the Royal College of Physicians and the Renal 2. Locatelli F, Alijama P, Barany P et al. Revised Association, and the Royal College of General European best practice guidelines for the manage- 56 Chapter 5 • Renal anaemia

ment of anaemia in patients with chronic renal Physicians of London and the Renal Association, failure. Nephrol Dial Transplant 2004; 19 (Suppl 2): 2002. 1–47. 5. Dreicher R, Horl WH. Differentiating factors 3. NICE. Anaemia management in people with between erythropoiesis-stimulating agents. Drugs chronic kidney disease. NICE Guideline. London: 2004; 64: 499–509. National Institute for Health and Clinical 6. Locatelli F, Aljama P, Barany P et al. Erythropoiesis- Excellence (NICE), 2006. stimulating agents and antibody-mediated pure 4. Renal Association. Adult Section 7: Anaemia in red-cell aplasia: where are we now and where do patients with chronic renal failure. In: Treatment of we go from here? Nephrol Dial Transplant 2004; 19: Adults and Children with Renal Failure: Standards and 288–293. Audit Measures, 3rd edn. London: Royal College of 6

Renal bone disease

Robert Bradley

Renal bone disease is one of the classic compli- glomerular filtration rate, GFR). The relevant cations associated with chronic kidney disease biochemical parameters are serum phosphate, (CKD). The normal physiological mechanisms serum calcium, calcium–phosphate product, regulating blood levels of phosphate, calcium, vitamin D levels and parathyroid hormone vitamin D and parathyroid hormone are dis- (PTH) levels. Each is important but it is the com- rupted and this has important implications for plex interaction between them that can be used the structural integrity and long-term health of to describe why ROD occurs. bone. It is now clear that the complex patho- The kidneys play a vital role in controlling physiology is not restricted to bone and that the each of the above parameters so virtually all increased risk of calcification, especially of the CKD patients, particularly those at stage 4 and cardiovascular system, is extremely important 5 CKD, will at some point exhibit a set of blood and probably has a role in the high cardio- results that include risk factors for ROD. See vascular morbidity and mortality in this popu- Chapter 4 for stages of CKD. lation of patients. Drug therapy with phosphate Phosphate is excreted by the renal tubules, so binders and vitamin D is the mainstay of treat- in the presence of a reduced GFR, urinary clear- ment. Patient education is crucial, particularly ance is reduced, leading to a build-up in the for phosphate binders, which must be taken blood (hyperphosphataemia). The normal range appropriately in relation to meals to demon- (e.g. 0.8–1.45 mmol/L) is frequently exceeded strate any benefit. Drug therapy can also exac- and values well in excess of 2 mmol/L are regu- erbate renal bone disease – hypercalcaemia or larly seen, particularly in the later stages of oversuppression of parathyroid hormone, for CKD. example, need to be avoided. Therapeutic Vitamin D is crucial for good bone health targets for the various parameters of bone because it promotes the gastrointestinal absorp- biochemistry are evolving, research into calcifi- tion of calcium, influences the renal tubular cation processes is ongoing and new pharmaco- reabsorption of calcium and aids the mineral- logical agents have recently become available. isation process in the bone. Vitamin D is pres- All of this will, it is hoped, improve the ent in the body because of the action of management of this complicated and serious sunlight on the skin and through dietary condition. absorption. This form of vitamin D is inactive (cholecalciferol) and requires enzymatic hy- droxylations in the kidney and the liver to become the active form of vitamin D, calcitriol Pathophysiology (1␣,25-dihydroxycholecalciferol). The renal reaction occurs under the control of 1␣- Renal bone disease (or renal osteodystrophy, hydroxylase – as renal mass reduces and enzyme ROD) is a breakdown in the homeostatic mech- levels fall there is reduced capacity for this vital anisms that control bone biochemistry, trig- stage of the process and activated vitamin D gered by a decline in renal function (the levels fall (Figure 6.1).

57 58 Chapter 6 • Renal bone disease

Action of sunlight Dietary intake

Cholecalciferol

Liver Kidney

CKD 1α-hydroxylase

25-hydroxycholecalciferol 1α-hydroxycholecalciferol (alfacalcidol)

1α,25-dihydroxycholecalciferol (calcitriol) Activated vitamin D

Promotes gastrointestinal Promotes bone calcium absorption mineralisation

Figure 6.1 Vitamin D metabolism in chronic kidney disease.

Under conditions of normal calcium homeo- CKD means that drug therapy should com- stasis, around half of the 1000 mg (25 mmol) of mence in CKD stage 4 and even stage 3 if indi- calcium that is generally ingested per day is cated by blood results, rather than delaying absorbed from the gut. The vitamin D deficit until stage 5 when the consequences of ROD and to a lesser extent the excess phosphate (via may already be established and drug manage- binding to free calcium in the blood) result in a ment is more difficult. fall in serum calcium below the normal range The parathyroids are a set of four small (e.g. 2.2–2.6 mmol/L) typically to just below glands situated, as their name suggests, in close 2 mmol/L. However, patients with CKD do not proximity to the thyroid gland. They secrete usually develop symptoms of hypocalcaemia. parathyroid hormone (PTH), which is funda- Acute changes to phosphate, calcium and mentally important for the maintenance of vitamin D as outlined above are unlikely to be normal bone turnover, and regulation of bone a problem for a CKD patient but left uncor- biochemistry. PTH secretion rises above the rected as CKD progresses they will increase basal level in response to hypocalcaemia, vita- the risk of developing ROD, especially as the min D deficiency and/or hyperphosphataemia. parathyroid glands begin to respond to the ab- PTH has a variety of mechanisms to restore normalities in bone biochemistry. normality, for example: In early CKD when the GFR may be well • Decrease urinary clearance of calcium above 25 mL/min small rises in phosphate are • Increase urinary clearance of phosphate seen as filtration becomes defective. Renal acti- • Increase synthesis of vitamin D vation of vitamin D also declines and the com- • Increase bone turnover to mobilise calcium bination of these two changes will reduce from bone to serum. calcium levels. As GFR continues to fall below 25 mL/min these abnormalities of bone mineral Under normal physiological conditions the homeostasis become more pronounced. actions of PTH will correct the bone biochem- However, the fact that this process starts early in istry and through negative feedback PTH secre- Pathophysiology 59

ȇGFR

ȇ ȇ urinary PO4 clearance Vitamin D synthesis

Ȇ ȇ serum PO4 serum Ca

Parathyroid glands

Ȇ CKD PTH secretion CKD

PTH actions on bone: PTH actions on kidney: ȆCa release? ȆCa retention X Ȇ Ȇ PO4 release? PO4 excretion X (CKD induces resistance ȆVitamin D synthesis X to bone actions of PTH)

Progression of CKD

Bone biochemistry profile remains abnormal Failure of negative feedback on parathyroid gland ȆȆPTH secretion

Hyperplasia of parathyroid tissue

Secondary hyperparathyroidism

Figure 6.2 Secondary hyperparathyroidism in chronic kidney disease.

tion then falls back to basal levels. In CKD the stimuli and PTH secretion rises well above the first three actions listed above will be impaired normal range (0.9–5.4 pmol/L). At a cellular because they rely on functioning renal tubules. level within the parathyroid tissue hyperplasia However the extensive reserve capacity of the will eventually occur and parathyroid glands kidney means that as long as GFR is above will enlarge. The high PTH secretion will con- 25 mL/min the response induced by PTH can be tinue to increase bone turnover, causing bone near to normal. The main focus is then to damage and release of phosphate from the increase serum calcium by the non-renal actions bone, exacerbating the hyperphosphataemia. of PTH on bone. This is unlikely to be sufficient The response of the glands to bone bio- to correct the calcium and meanwhile phos- chemistry is now pathological and secondary phate retention continues and vitamin D syn- hyperparathyroidism (HPT) is present (Figure thesis falls as GFR continues to drop. The 6.2). This complication is present to some negative feedback system now fails, the para- degree in the majority of stage 5 CKD patients thyroid glands continue to respond to these on regular dialysis therapy. 60 Chapter 6 • Renal bone disease

Complications for the skeleton and This is referred to as osteitis fibrosa and is the progression of renal bone disease most common and important form of ROD. Osteopenia is a condition of bone that occurs when bone resorption exceeds bone formation The complex bone biochemistry abnormalities from whatever the cause. On radiographs this that initiate renal bone disease result in equally appears as increased radiolucency or ‘poverty of complex changes to the skeleton, especially bone’. Osteopenia can be associated with relating to rate of bone turnover (Figure 6.3). osteitis fibrosa and also with hypogonadism. For Vitamin D deficiency may be associated with a variety of reasons, gonadal steroid hormone low bone turnover. Osteoblasts (bone formation levels are low in both men and women with cells) and osteoclasts (bone resorption cells) more advanced stages of CKD. need vitamin D to function effectively. Without It is important to note that the pathogenesis it, bone mineralisation is defective and osteoid of bone disease in CKD is a series of complicated (immature bone that has not yet undergone cal- processes and multifactorial in origin. Many cification) accumulates in the bone. This is patients will have mixed ROD, exhibiting bone known as osteomalacia and is associated with damage secondary to both low and high significant damage to the structural integrity of turnover. The specific type of bone disease that bone. There are other forms of low bone a patient has can only be confirmed on bone turnover states, some of which are related to biopsy. In practice this is a painful, invasive treatment options in ROD, and will be described intervention that is rarely carried out. Drug later. therapy is based on the relative degree of abnor- Elevated PTH levels are associated with high mality of the bone biochemistry profile and on bone turnover. There is increased remodelling of general principles to promote good bone health. bone with greater activity of osteoclasts relative A raised bone specific alkaline phosphatase to osteoblasts. This weakens the bone architec- level, indicative of bone damage, is often seen, ture and induces fibrotic changes in the bone. particularly in the presence of osteitis fibrosa.

Secondary hyperparathyroidism Vitamin D deficiency

Ȇosteoblast activity ȇosteoblast activity ȆȆosteoclast activity ȇȇosteoclast activity defective mineralisation

high bone low bone turnover turnover

osteitis mixed osteomalacia fibrosa renal osteodystrophy

pathological fractures microfractures bone pain muscle pain

Figure 6.3 Bone diseases in chronic kidney disease. Complications for the skeleton and renal bone disease 61

The clinical consequences of all the effects of • Exacerbation of anaemia CKD and HPT on bone are to increase the risk • Soft tissue calcification of pathological fractures (e.g. vertebral frac- • Peripheral vascular calcification tures), bone pain, muscle pain, microfractures • Cardiovascular calcification. and even skeletal deformities.1,2 In spite of pharmacological intervention, Hyperphosphataemia is primarily responsible bone biochemistry abnormalities may persist for pruritis and red, sore eyes. Itching is a com- and the secondary HPT progresses as the mon problem for CKD patients and is related parathyroid glands continue to be overworked. to their uraemic state. It is one of the most This results in: troublesome and distressing symptoms and management of high phosphate can contribute • Further increases in PTH levels (can reach to symptom relief. 8–10 times the normal range or more) Hyperparathyroidism may cause bone mar- • Increased hyperplasia of parathyroid glands row fibrosis which can contribute to the sub- • Reduced vitamin D receptors on the surface optimal haemoglobin of renal anaemia and be of the glands a factor in poor response to therapy with • Reduced sensitivity of parathyroid tissue to erythropoiesis-stimulating agents (ESAs). the suppressive effects of calcium. Soft tissue, peripheral vascular and cardio- Over time, the parathyroid glands become vascular calcification are particularly important 3 less responsive while they continue to secrete and are the focus of much research at present. more and more PTH. Bone turnover accelerates, Calcium and phosphate remain soluble in the worsening the osteitis fibrosa. More calcium and blood under normal physiological conditions phosphate move from the bone to the serum and there are various factors that maintain the under the influence of PTH, exacerbating hyper- status quo. If serum levels of calcium, phos- phosphataemia and increasing calcium levels. phate or both rise, there is a risk that the With advanced HPT the parathyroid gland has concentrations can reach a level where precipi- greatly reduced sensitivity to calcium levels tation occurs, resulting in extraskeletal calcifica- because there are fewer calcium-sensing recep- tion. It is clear that CKD patients have potential tors on the surface of the gland. The set point risk factors for calcification, for example: at which calcium effects PTH secretion is raised. • Hyperphosphataemia So as the PTH remains high, the serum calcium • Hypercalcaemia in the presence of severe will eventually rise above the normal range. HPT This stage of ROD is tertiary HPT and the glands • Hypercalcaemia related to drug therapy for are acting autonomously. ROD (e.g. vitamin D and calcium-based Clearly, pharmacological management now phosphate binders, to be described later) becomes increasingly difficult, the parathyroid • High calcium–phosphate product glands become increasingly resistant to the • Vitamin D deficiency leading to poor calcium action of drugs and surgical options (i.e. para- utilisation. thyroidectomy) may need to be considered. It also explains why the calcium–phosphate product is an important parameter to monitor Extraskeletal complications in CKD patients; it is calculated simply by mul- tiplying the serum concentration of calcium by It is becoming increasingly apparent that there that of phosphate and measured in mmol2/L2 are other consequences of the bone biochemical (Figure 6.4). changes that are extremely important for CKD Soft tissue calcification involving the periph- patients. eral arteries can develop into a rare but severe Other potential complications include: condition called calciphylaxis or calcific • Pruritis uraemic arteriolopathy. This is characterised by • Red, sore eyes tissue and skin ischaemia leading to severe 62 Chapter 6 • Renal bone disease

CKD progression? ROD treatment failure:- Poor compliance? Delayed treatment?

ȆȆ ȆȆ ǟǞȆ PO4 PTH Ca Calcification risk Parathyroid gland changes:- Further hyperplasia ȇ Ca sensing receptors Severe secondary HPT ȇVitamin D receptors

Further bone damage ȆȆBone turnover Impaired capacity to

Standard drug therapy:- “buffer” PO4 and Ca Vitamin D ǞȆCa ǞȆ ȆȆȆ ȆȆȆ PO4 PO4 Ca Calcification risk

Calcium based PO4 binders ǞȆCa

Tertiary (hypercalcaemic) HPT

ȆȆȆȆ ȆȆ × ȆȆ ȆȆȆ PO4 Ca Ca PO4 product PTH

ȆȆȆCalcification risk

Soft Peripheral Coronary Cardiac tissue arteries arteries valves

Figure 6.4 Progression of bone biochemistry abnormalities, hyperparathyroidism and extraskeletal calcification risk in chronic kidney disease. necrosis. It is an extremely painful complication cardiac valves) is an extremely important issue with a very high mortality rate, usually because for patients with CKD. It is well established that, of sepsis related to the necrotic skin lesions. in the end stage renal disease (ESRD) popula- Metastatic calcification (of which calciphy- tion, the most common cause of death is of laxis is an extreme form) can also occur in the cardiovascular origin. For example, the cardio- lungs, cornea, conjunctivae, muscle, kidney and vascular mortality of dialysis patients is on aver- stomach. Simple but painful skin or periarticu- age 30 times that of the general population.4 lar deposition of calcium may be visible in CKD This figure is even higher when data are patients. restricted to younger patients. Nearly half of all Calcification in the cardiovascular system deaths in dialysis patients can be attributed to (e.g. myocardium, coronary arteries, aorta and cardiovascular disease.5 Disturbances to bone Treatment with phosphate binders 63 mineral homeostasis are increasingly being Treatment with phosphate binders implicated in the high mortality rates. The exact role of calcification in the high Managing hyperphosphataemia can be ex- cardiovascular morbidity and mortality in CKD tremely difficult in CKD patients. Many of the patients is yet to be fully determined. One pharmacological therapies used are poorly toler- hypothesis is that vascular smooth muscle cells ated, and efficacy is totally reliant on the are triggered by certain factors to undergo a patient taking the prescribed agent at specific change in phenotype to become bone-forming times. Phosphate binder drugs act to reduce the (osteoblast type) cells. It will be crucial to estab- absorption of dietary phosphate so they must be lish the precise nature of the pathology of vas- taken either with or just before meals and so are cular calcification and whether drug therapy for plagued by concordance problems. All binders ROD can prevent or reverse cardiovascular form a complex with phosphate in the gut calcification. The roles of other classical cardio- which is not absorbed systemically and will be vascular risks such as atherosclerosis related to cleared from the body via the faecal route. By dyslipidaemia, chronic inflammation, hyperten- reducing the gastrointestinal absorption the sion and diabetes need to be elucidated to com- blood levels are not continually topped up, plete the calcification jigsaw in CKD patients. allowing them to fall gradually. A consequence of arterial calcification is that Taking phosphate binders inappropriately, vessels such as the aorta and coronary arteries for example between meals, will have little become stiff. Left ventricular hypertrophy, effect on phosphate. This can lead to doses of reduced coronary artery blood flow during the binders being increased to address the elevated diastolic phase and higher systolic blood pres- phosphate when a simple piece of patient edu- sure are all potential complications of greater cation would address the problem while main- arterial stiffness and reduced vascular compli- taining the same dose. For this reason renal ance. Valvular calcification of the aortic or healthcare professionals, including pharmacists, mitral valves can also occur and may contribute have an important role to play in talking to to heart failure. patients with CKD about the benefits of phos- Epidemiological studies provide data on the phate binders and how to take them correctly. roles of abnormal phosphate, calcium and PTH Also, although phosphate binders are generally values as risk factors for mortality in patients prescribed as a three times a day dosage, with ESRD.6 Retrospective data in patients on patients should be encouraged to adjust their haemodialysis have indicated a twofold mortality doses according to their diet. For example, only rise with serum phosphate above 2.26 mmol/L.7 two doses are required if the patient has only A similar retrospective study demonstrated a two meals a day and additional, smaller doses of 27% higher mortality risk, adjusted for age and binders should be taken with snacks. co-morbidities, in patients on haemodialysis with The present range of available phosphate a phosphate over 2.10 mmol/L.8 Even at lower binders fall into three categories: calcium-based, phosphate levels than this the risk is still high aluminium-based and calcium/aluminium-free and unfortunately, due to the difficulties in agents. managing phosphate, around 50% of dialysis patients worldwide are estimated to have serum phosphate levels above 1.8 mmol/L.9 Calcium-based phosphate binders Hyperparathyroidism may have additional unfavourable effects on the cardiovascular • Calcium carbonate (e.g. Calcichew, 500 mg system, such as myocardial interstitial fibrosis elemental calcium per tablet, chew 5–10 min- and thickening of arteriolar walls. Following on utes before meals) from this it is vital to elucidate if and to what • Calcium acetate (e.g. Phosex, 250 mg ele- extent modification of the risk factors related to mental calcium per tablet, swallow whole ROD affect cardiovascular outcomes. with meals). 64 Chapter 6 • Renal bone disease

This group are the most widely prescribed, and toxic to the bone marrow (so may worsen renal will be first-line agents for most patients as they anaemia) and to bone cells, causing greatly are cheap and relatively efficacious. Calcium reduced bone turnover (adynamic bone disease). binds to phosphate in the gut and the resulting A direct link between aluminium-based binders complex is not absorbed. Some of the calcium and these complications is not clear. Although that remains unbound will be absorbed, which serum aluminium levels can be monitored, their is desirable in hypocalcaemic patients but limits clinical usefulness is debated and there is no or contraindicates their use in patients with threshold level for aluminium-induced dis- more severe forms of HPT. The starting dose is eases.11 The starting dose of these agents is one one or two tablets at mealtimes. It is important capsule with each meal and the main indication that patients understand when to take them as is as a second- or third-line agent or as an add- doses being taken between meals act as a cal- on therapy when a single agent has not ade- cium supplement, with maximum absorption. quately reduced phosphate levels. To minimise Concordance issues include gastrointestinal dis- toxicity, aluminium-based binders are used only turbances such as nausea and poor palatability, on a short-term basis. As with all phosphate since the chewable formulations have a chalky binders, gastrointestinal intolerance, in this case texture. There is conflicting evidence concern- constipation, is a problem. ing the relative merits of Calcichew and Phosex on a milligram for milligram of elemental calcium basis with regard to effectiveness of Calcium/aluminium-free phosphate binders binding gut phosphate and incidence of hyper- calcaemia. The efficacy of calcium salts as phos- • Sevelamer hydrochloride (Renagel, 800 mg phate binders is influenced by gut pH levels, so tablets, swallow whole with meals). could be modified by acid suppression therapy, although this appears to be less of a problem Sevelamer is the newest phosphate binder avail- with Phosex. able and is a completely new class of drug. It is a hydrogel of poly(allyamine hydrochloride), a polymer molecule with partially protonated Aluminium-based phosphate binders amine groups which bind to intestinal phos- phate. Its advantages are that, being free of calcium, it is safer to prescribe for patients • Aluminium hydroxide (e.g. Alu-Cap capsules, who are hypercalcaemic or in those with evi- 475 mg capsules, swallow whole with meals). dence of calcification. Sevelamer has been These agents are the most potent phosphate shown to have a lower incidence of hyper- binders currently available.10 Concerns over calcaemia than calcium-based binders.12 It is toxicity of absorbed aluminium, especially also aluminium-free so is a viable option when dementia, led to a reduction in their use. aluminium toxicity is a concern. Unbound However the elevated serum aluminium levels sevelamer is not absorbed from the gut so it has seen in patients on haemodialysis were more no systemic side-effects but still has gastro- likely to be related to absorption of the metal intestinal side-effects such as nausea. It is prob- from the water used in the haemodialysis ably the least effective of the current range of process. Water purification requirements are drugs, which can cause a problem with high now more stringent and most aluminium is tablet burden. There is some concern about risk removed before it reaches the haemodialysis of exacerbating metabolic acidosis in patients machine. Current evidence that phosphate with CKD.13 Cost is also an issue because should be controlled more aggressively to sevelamer is much more expensive than tradi- improve patient outcomes has led many renal tional therapies. It is important to be able to jus- units to increase prescribing of Alu-Cap capsules tify its prescription on sound clinical grounds so once more. However there are other significant that sevelamer can be utilised in a cost-effective adverse reactions linked to aluminium. It is manner. Treatment with vitamin D 65

An interesting effect of sevelamer is an action phate may have less impact on serum levels. to bind bile acids in the gut, leading to a reduc- Non-pharmacological management of hyper- tion in serum LDL (low-density lipoprotein) phosphataemia revolves around dietary modifi- cholesterol by up to 20%. This may have bene- cation. The issue of reducing phosphate intake fits in terms of reducing cardiovascular risk in the diet is controversial. Many foods contain factors in patients with CKD.14 phosphate; dairy products and protein-rich The starting dose of sevelamer is one or two foods such as meat are especially rich and would tablets with meals although doses of up to five be restricted in a low-phosphate diet. However, tablets with each meal may be required. It is nutrition in general can be a major problem for generally prescribed as a second- or third-line renal patients and a low-phosphate diet is pro- agent, or as an additional therapy in combina- tein and calorie poor. For this reason many tion with calcium- or aluminium-based binders. nephrologists and dieticians advocate a nutri- There are novel phosphate binders in clinical tious diet with sufficient protein rather than development or new to the market (e.g. lan- phosphate restriction. It is then the role of thanum carbonate (Fosrenal)) but any di- or phosphate binders to regulate serum phosphate. trivalent cation will bind phosphate. Iron- and For patients with ESRD, treatment with stan- magnesium-based compounds, for example, dard techniques of haemodialysis or peritoneal have been used as phosphate binders. dialysis has little impact on phosphate clearance Phosphate binders are chronic therapies pro- because the majority of body phosphate is intra- ducing gradual changes in the bone bio- cellular so that clearance is slow. More frequent chemistry so dose adjustments are usually not and longer than usual haemodialysis sessions made more frequently than every two to four can help to reduce doses of phosphate binders. weeks during initial phase of treatment and then every four to eight weeks thereafter. Crucially, reduction in phosphate levels can Treatment with vitamin D provide a directly associated suppression of PTH secretion and, of less significance, an increase in free calcium levels in the serum. Gastro- Vitamin D therapy works in conjunction with intestinal intolerance and issues around con- phosphate binder drugs and most renal patients cordance and patient education are the most will be prescribed it at some point. As CKD problematic issues with all of the phosphate progresses, the quantity of activated vitamin D binders. produced will continue to fall, leading to the Combination therapy with different phos- complications outlined earlier. Levels of vitamin phate binders may sometimes be seen in clinical D in the blood can be measured and this can practice. This could be indicated in cases of guide its use as a supplement, particularly in the recalcitrant hyperphosphataemia, but in situ- early stages. However, in most renal units vita- ations like this the combination is often min D is used more as a pharmacological agent intended to be short term to gain better control than as a simple supplement, and blood levels of phosphate. It is important to review such are rarely checked. The prescription and dose patients regularly to avoid polypharmacy and adjustment of vitamin D is based on other increasing the complexity of the drug regimen parameters of ROD, namely calcium and PTH, unnecessarily. Long-term therapy with two or to control secondary HPT. The initiation of vita- more binders in high doses is rarely indicated – min D is often in response to a low calcium an elevated phosphate despite such intense level but ideally a PTH value should also be therapy can suggest either a problem with con- available because, once the calcium is corrected, cordance or the presence of severe bone disease. it is the PTH level that is more likely to guide At the extremes of bone turnover states (ady- subsequent dose adjustments in the long term. namic or severe hyperparathyroid) the bone Moreover, unless the serum calcium and PTH loses much of its ability to buffer bone minerals are particularly disrupted, prescription of phos- so reduction in the absorption of dietary phos- phate binders alone in the first instance will 66 Chapter 6 • Renal bone disease often increase calcium and reduce PTH, delay- times a week are often given on haemodialysis ing the need for vitamin D. units. While individual patients may obtain Vitamin D supplementation will prevent better control of their PTH, there is not a large osteomalacia and has both indirect and direct evidence base to demonstrate superior efficacy actions in treating secondary HPT. Gastro- over standard daily dosing.15 However, this intestinal absorption of calcium is promoted, approach does have the advantage of guaran- increasing the serum calcium. This in turn stimu- teeing concordance with treatment. lates the calcium-sensing receptors on the surface Alfacalcidol and calcitriol are also available as of the parathyroid gland, inducing a drop in PTH intravenous preparations for pulsed treatment. secretion. Vitamin D also has a direct effect, sup- However the benefits of this practice over tradi- pressing the hormone by inhibiting PTH gene tional oral therapy regimens have not been transcription in the parathyroid gland cells. clearly demonstrated.16 There is also the added There are two forms of vitamin D used in factor of greatly increased price compared with patients with CKD: the oral formulations. Low bone turnover as a • Alfacalcidol (1␣-hydroxycholecalciferol), consequence of excessive vitamin D dosage also usual starting dose 0.25 µg once a day reduces the buffering action of bone on phos- • Calcitriol (1␣,25-dihydroxycholecalciferol), phate and calcium levels because the under- usual starting dose 0.25 µg once a day. active bone will not take up these minerals, increasing the risk of them being deposited else- Alfacalcidol still requires activation in the liver, where as ectopic calcification. whereas calcitriol is the active form of vitamin For dialysis patients, adjusting the calcium D. Both agents are effective at managing content of their dialysis fluids can be a non- secondary HPT but alfacalcidol is more widely pharmacological method of improving ROD prescribed in the UK. Calcium levels can be cor- management. Careful reduction of the calcium rected relatively quickly but PTH correction is a content, for example down to 1 mmol/L will more gradual process. For this reason, vitamin D reduce the net positive calcium burden to which therapy is adjusted infrequently, every 4–12 patients with CKD are frequently subjected. This weeks in most instances. reduces the risks of hypercalcaemia, allowing The clinically significant side-effects of vita- greater doses of vitamin D to be employed min D therapy relate to inappropriate changes where necessary to control HPT (Figure 6.5). to the bone biochemistry: • Exacerbation of hyperphosphataemia (via promotion of phosphate absorption from the The role of parathyroidectomy gut) • Hypercalcaemia • Oversuppression of parathyroid glands (PTH For some patients, surgical intervention is even- is essential for normal bone turnover so over tually necessary to manage the complications zealous treatment with vitamin D can reduce associated with ROD. A parathyroidectomy may PTH to a level at which the patient has a risk be indicated in a number of circumstances, for of adynamic bone disease). example: The first two complications, especially hyper- • Presence of tertiary HPT with symptomatic calcaemia, can limit the efficacy of vitamin D hypercalcaemia that has not responded to therapy. The degree of secondary HPT often dic- avoidance of calcium-based phosphate tates a dose increase but the presence of raised binders and minimisation of vitamin D dose. calcium can preclude this. Various strategies are • Resistant HPT that has not responded to high employed in an attempt to maximise the effects doses of vitamin D and phosphate binders. of vitamin D on the parathyroids while limiting This could be severe disease that has not its action on the gut vitamin D receptors. Pulsed been adequately treated or it could be related doses of vitamin D, given once, twice or three to poor concordance with drug therapy. As The role of parathyroidectomy 67

Vitamin D induced Aluminium related PTH oversuppression Parathyroidectomy toxicity

PTH below range Osteomalacia set for CKD patients

ȇbone turnover

ȇosteoblast activity ȇosteoclast activity

Adynamic bone disease Impaired capacity to

‘buffer’ PO4 and Ca

Pathological fractures Microfractures Ȇ Can exacerbate PO4 Bone pain Ȇ Ȇ Ca and Ca PO4 product Muscle pain

Can add to the risk of extraskeletal calcification

Figure 6.5 Risk factors for low bone turnover states in chronic kidney disease and associated complications.

with the majority of patients considered for There are three surgical options: parathyroidectomy PTH levels will be very • Subtotal parathyroidectomy high – typically 10 times above the normal • Total parathyroidectomy reference or higher. • Total parathyroidectomy with reimplanta- • Patients with adenomas of the parathyroid tion. glands. These are the more extreme hyper- Removal of all four glands in a total para- plastic manifestation of HPT, following on thyroidectomy will leave the patient devoid from diffuse then nodular hyperplasia of the of PTH. While this may address the problems gland tissue. They are generally resistant to that led to the surgery it can present new ones. drug therapy due to greatly reduced concen- With no PTH to regulate normal bone cell bio- trations of both vitamin D and calcium-sens- chemistry, adynamic bone disease can result, ing receptors on the surface of these nodules which itself carries an increased risk of frac- compared with normal parathyroid tissue. tures. For this reason some parathyroid tissue is • Patients with evidence of significant sequelae often left behind either at the site of the of abnormal bone biochemistry, such as cal- parathyroid glands (subtotal parathyroid- ciphylaxis and severe bone damage. ectomy) or removed from this site and placed 68 Chapter 6 • Renal bone disease in the forearm (total parathyroidectomy with the massive burden of cardiovascular disease reimplantation). The resultant risk from either in the CKD population by preventing cardio- of these techniques is that the remaining vascular calcification. parathyroid tissue is still sufficient to cause sig- In order for these outcomes to be realised, it nificant HPT, necessitating further surgery. The is essential that the target ranges for each of the latter technique has the advantage of leaving parameters of bone biochemistry are clearly parathyroid tissue in a more accessible site for defined and that guidance is available on the subsequent operations. most effective pharmacological means to Symptomatic hypocalcaemia is a significant achieve them. In ROD this is crucial, as there are risk in the early post-parathyroidectomy phase different approaches to drug management and, and left uncorrected can progress to hypocal- more importantly, it is not simply a question of caemic tetany. The risk is higher for total returning the serum levels of phosphate, cal- parathyroidectomy and in those patients not cium and PTH to the normal reference ranges. prescribed a short course of high-dose vitamin The evidence base currently available on these D pre-operatively. Serum calcium should be issues is variously regarded as incomplete, managed aggressively when it begins to drop inconclusive or conflicting and is complicated after surgery, using high doses of vitamin D by emerging data surrounding cardiovascular (sometimes in the region of 4 µg/day) and cal- calcification. Evidence-based guidance on ROD cium supplements (given as large oral doses or is published by nephrology groups such as the intravenously if indicated). Calcium carbonate Renal Association in the UK17 and the National (e.g. Calcichew) is often used but has the poten- Kidney Foundation in the USA.18 The advice tial to cause confusion for the patient. They may vary depending on the stage of CKD that may have taken Calcichew as a phosphate the patient has reached. The following descrip- binder in the past at mealtimes. They must be tions of bone biochemistry targets include ref- informed that it now serves as a calcium sup- erence to these guidelines and are generally plement and should take it between meals to applicable to the later stages of CKD. Current maximise absorption. Vitamin D therapy and recommendations are different from those of 5 possibly calcium replacement may be needed or 10 years ago. It is worth remembering that in long term, but the risk and magnitude of an area such as this where new data are appear- hypocalcaemia diminishes over time and doses ing regularly, they could be amended once more of these drugs should be reviewed regularly to in the near future. avoid hypercalcaemia.

Phosphate Therapeutic targets For the majority of patients with CKD it is extremely unlikely that a normal serum phos- The benefits of drug therapy to manage ROD phate will be achieved with binder drugs. In may not be immediately obvious to the patient fact, hypophosphataemia in a patient with CKD and it is important that they understand their indicates malnourishment and is an indicator of therapy. Phosphate binders and vitamin D are poor prognosis. Vitamin D therapy, a normal or long-term treatments that will gradually correct high-phosphate diet, severe HPT and poor con- abnormalities in their blood results, protect the cordance with binder drugs are some of the fac- bones and possibly the heart and blood vessels tors that promote hyperphosphataemia and against future damage. Successful drug therapy make successful management very difficult. A reduces the morbidity and mortality associated more realistic, though still difficult, objective is with ROD by reducing fracture incidence, and to bring serum phosphate down below 1.7–1.8 preventing soft tissue calcification. As the mmol/L and this should have a positive effect emerging evidence indicates, it also reduces on patient mortality rates. New management strategies 69

Calcium secretion is necessary19 and target levels need to be higher than normal, but to what degree is a Depending on the stage of ROD, either hypo- or crucial question. Current theories indicate that hypercalcaemia may be present, so ideally drug a value between three and five times the upper treatment should maintain a calcium level in limit of normal should be aimed for in patients the normal range. Current concerns about cal- with ESRD. This should manage the HPT effec- cification risks have led some nephrologists to tively while avoiding oversuppression of the advocate a serum calcium towards the lower end parathyroid glands and maintaining as near as of normal. possible a normal bone turnover state. The incidence of low turnover bone disease has been increasing, partly because of excessive PTH sup- Calcium–phosphate product pression with vitamin D, and this is reflected in the changes to the PTH target range. However, Guidance is variable but most suggest a recent worldwide data indicate that around calcium–phosphate product in the region of less 50% of dialysis patients have a PTH below three than 4.50 mmol2/L2 to minimise calcification times the upper limit of normal (less than risks. Aiming to keep the phosphate below 16.2 pmol/L),9 suggesting that a change in prac- 1.7 mmol/L and the calcium in the lower end tice is required. of the normal range will assist in the achieve- ment of target calcium–phosphate product. Current data indicate that around 50% of dia- lysis patients have a product greater than New management strategies and 4.44 mmol2/L2.9 new therapeutic agents for renal bone disease

Parathyroid hormone Phosphate management An important factor to consider with PTH tar- gets is that levels in the normal range are no One of the current controversies in ROD is the longer viewed as desirable in most patients with role of calcium intake in the pathogenesis of CKD. As described earlier, PTH is a vital com- extraskeletal, especially cardiovascular, calcifica- ponent in the physiology of normal bone tion. CKD patients have the potential to be in turnover. Patients with CKD, especially those on positive calcium balance, both through haemodialysis, are often described as being in a enhanced gastrointestinal absorption of exo- proinflammatory state and this may be reflected genous calcium from calcium-based phosphate in elevated levels of inflammatory markers such binders and due to vitamin D therapy. as CRP (C-reactive protein). Inflammatory Mineralisation of the bone is defective in CKD cytokines (e.g. interleukins 1, 6 and 11 and tis- and the action of elevated PTH will mobilise cal- sue necrosis factor alpha) can increase bone cium from the bone. Both of these factors mean remodelling. Also, bone growth factors may be that normal physiological utilisation of calcium deficient or inhibited in CKD and suppressors of is impaired. The next question is, where does bone formation may be present at higher levels the positive balance of calcium go if it is not than in the general population. Finally, there taken up by the bone? Does its utilisation may be a relative resistance to the actions of become pathological, with exogenous calcium PTH in CKD, possibly due to downregulation contributing to extraskeletal calcification? Also, of PTH receptors. Endogenous factors are what is the role of ingested calcium in the cell- induced to attempt to overcome these changes ular processes underpinning arterial calcifica- and normalise bone turnover. In CKD, PTH is tion? Research is ongoing in these areas and one of these factors so a degree of hyper- more data are needed, especially since opinion 70 Chapter 6 • Renal bone disease is polarised in the nephrology field as to the given to a patient with CKD – for example in risks and benefits of prescribing calcium-based younger patients who face years of phosphate phosphate binders. binder ingestion, in an effort to avoid or delay One problem for clinicians assessing calcifi- calcification. An evidence base needs to be cation risk is that of the approximately 1000 g firmly established to compare sevelamer and of calcium present in the body, serum calcium calcium-based binders with respect to risk of represents only a tiny proportion of total body developing cardiovascular calcification, rate of calcium (approximately 0.025%). Around 99% progression of cardiovascular calcification and, of the total body calcium is present in bone. crucially, cardiovascular morbidity and mortal- Does serum calcium reflect total body calcium ity. Sevelamer has been compared with calcium- sufficiently well to indicate calcification risk? If based phosphate binders after 12 months not, then could a patient with normal serum therapy in dialysis patients and elicited an calcium still be at risk and could the risk be attenuation in cardiovascular calcification as enhanced by adding to the patient’s calcium assessed by EBCT score not seen with the load with calcium-based phosphate binders? calcium-based products.22 Various techniques such as echocardiography Lanthanum carbonate (Fosrenol) is a new can visualise calcification to some degree, but agent that has just received a European licence. obviously in these patients the complication has Lanthanum is a rare earth element that binds already manifested itself. A newer technique dietary phosphate so will be prescribed in the called EBCT (electron beam computed tomo- same manner as the current range of drugs. graphy) can have a role in assessment of Early data suggest that potency could be similar coronary artery calcification by providing a cal- to that of aluminium preparations. cification score, which can then be matched to Whatever phosphate binder is used, the ac- a level of cardiovascular risk.20 EBCT has been cumulating evidence that high serum phos- used to compare young patients (20–30 years of phate levels are linked to increased mortality in age, many of them taking calcium-based patients with CKD (seemingly more so than binders) on haemodialysis with non-dialysis other measures of bone mineral homeostasis patients with cardiac disease. Coronary artery like serum calcium and PTH) means that earlier calcification scores were found to be far higher and tighter control is necessary to achieve tar- in the former group.21 However, EBCT is unable get levels and improve outcomes for patients. to differentiate accurately between intimal and medial vascular calcification, making it difficult to assign a cause to the calcium deposition. This New vitamin D analogues is important because the former is more closely associated with the traditional cardiovascular There is a range of alternatives to alfacalcidol risk factor of atherosclerosis than the latter, and calcitriol either in use or in development. which may have aetiologies more particular to The rationale behind them is to overcome the CKD. dose-limiting side-effects associated with stan- All these factors may in future have a bear- dard vitamin D therapy. They purportedly do ing on the prescribing of phosphate binders. this through increased selectivity for the Will clinicians continue in the traditional man- parathyroid glands (to maximise PTH suppres- ner using calcium-based agents as first-choice sion) while sparing the actions on the gut vita- phosphate binders in most instances or will the min D receptors (to minimise the absorption of use of calcium-free phosphate binders as first- calcium). Of these vitamin D analogues only line drugs become more commonplace? This paricalcitol (Zemplar) is currently available in could be aluminium-based but is far more likely the UK. As with many aspects of drug therapy to be sevelamer. Many nephrologists are already for ROD there is debate about the role of these using sevelamer at an earlier stage than previ- new options. However at present there appears ously or even as the first phosphate binder to be an absence of good clinical trial data that New management strategies 71 demonstrate significant advantages in terms of Calcimimetics better efficacy or adverse event rate of the new agents over the current management strategies Cinacalcet (Mimpara) is the first of a new class with alfacalcidol or calcitriol.23 of agents called calcimimetics, offering a new Moreover, many of the new vitamin D ana- approach to the management of ROD. logues are only available as intravenous formu- Cinacalcet is an orally active compound which lations, effectively limiting them to patients on acts on the calcium-sensing receptors on the haemodialysis only, and they are much more surface of the parathyroid gland cells, mimick- expensive than existing options. ing the action of calcium. This induces a rapid Vitamin D receptors are distributed widely reduction in PTH secretion, offering a useful around the body so the effects of alfacalcidol, addition to the range of therapeutic options in calcitriol and the newer vitamin D analogues are the management of HPT. not restricted to the bone. Examples of postu- Cinacalcet will generally be prescribed in lated extraskeletal actions include: addition to rather than instead of phosphate binders and vitamin D. However, the associated • Inhibition of renin production and associated effects of calcimimetics (reductions in serum reduced angiotensin II levels, which indicates levels of calcium and phosphate) will be of a role in blood pressure homeostasis. benefit to many CKD patients and may allow • Possible protective role against mediators of for phosphate binder dose reductions, and more myocardial damage. importantly, aggressive use of concurrent vita- • Action on the pancreas to facilitate appropri- min D if required to suppress PTH. Patients with ate secretion of insulin. adenomas of the parathyroid glands have a • Immunomodulatory effects on various white deficit of calcium-sensing receptors so are blood cells. probably less likely to respond to this new • Anti-proliferative actions, which could be therapeutic option. At present due to its beneficial for the cardiovascular risk and pro- high cost, cinacalcet is restricted to the more tect against malignancy. difficult cases of ROD, for example those with These and other vitamin D-mediated effects severe HPT resistant to standard treatment, are currently under investigation and if con- those with HPT complicated by hypercal- firmed would suggest a significant and wide- caemia or as a pharmacological alternative to spread physiological role for vitamin D. parathyroidectomy.

C CASE STUDY

Mrs A is 45 years old and has CKD secondary to hypertension. She has been reviewed in the nephrology clinic and her renal function has now deteriorated from stage 3 to stage 4 CKD.

Blood results: • Phosphate 1.99 mmol/L (0.8–1.45 mmol/L) • Corrected calcium 2.10 mmol/L (2.2–2.6 mmol/L).

(continued overleaf) 72 Chapter 6 • Renal bone disease

C CASE STUDY (continued)

At her last clinic visit three months ago her phosphate was 1.65 mmol/L and corrected calcium 2.28 mmol/L.

Q1. Why is this patient’s hyperphosphataemia worsening?

Q2. Are there any symptoms she may now experience secondary to her elevated phosphate? A decision is made to manage the elevated serum phosphate with phosphate binder drugs.

Q3. What would you advise as first-line phosphate-binding agent for Mrs A? What dose would you recommend? How would you educate the patient on why phosphate binders are impor- tant, how they work, when to take them and what side-effects to expect?

Q4. What do you think is a realistic target serum phosphate level for Mrs A? A week after the nephrology clinic the junior doctor who reviewed Mrs A is looking for more advice. The patient was commenced on Calcichew (calcium carbonate tablets 500 mg elemental calcium per tablet) at a dose of two tablets three times a day as a phosphate binder, but the doc- tor is wondering whether vitamin D therapy should also be prescribed (blood results from clinic – phosphate 1.99 mmol/L and corrected calcium 2.10 mmol/L).

Q5. Is vitamin D therapy indicated for Mrs A at this point? If yes, which vitamin D agent and what starting dose would you recommend? If no, why do you think it is appropriate to leave her on Calcichew alone at present?

Q6. What other biochemical investigation(s) would you request at this stage in the management of Mrs A’s renal bone disease? How would knowledge of the result(s) influence your deci- sion to initiate or hold off vitamin D therapy? Three years have now passed by and you encounter Mrs A once again. She is now an ESRD patient on regular haemodialysis three times a week. Her current renal bone disease drugs are: • Calcichew one tablet three times a day • Alfacalcidol 0.5 µg once a day. This drug regimen has been stable for the past three months. Her current bone biochemistry profile is: • Phosphate 2.3 mmol/L • Calcium (corrected) 2.7 mmol/L • Parathyroid hormone (PTH) 52 pmol/L (0.9–5.4 pmol/L). During the last three months, phosphate has been stable but both calcium and PTH have gradu- ally increased. Mrs A reports that she has been experiencing more muscular aches and pains recently. She concords with her drug therapy and takes her Calcichew appropriately but in view of the mild

→ New management strategies 73

C CASE STUDY (continued)

(asymptomatic) hypercalcaemia, the nephrology team has decided to stop the Calcichew. Some expert advice is once again needed.

Q7. What are the other two commonly prescribed phosphate binders available to the nephrol- ogy team?

Q8. Considering the advantages and disadvantages of each of these agents, which one would you recommend and what would be your suggested starting dose?

Q9. How can vitamin D therapy cause complications in the management of renal osteodys- trophy?

Q10. What would you suggest as a vitamin D plan in Mrs A and what are the reasons behind your decision? Stop alfacalcidol? Reduce alfacalcidol dose? Continue alfacalcidol at current dose? Increase alfacalcidol dose? Later in the year, Mrs A is admitted to the orthopaedic ward with a fractured wrist following a fall. Her bone biochemistry profile is now: • Phosphate 2.1 mmol/L • Calcium (corrected) 2.6 mmol/L • PTH 69 pmol/L. An alkaline phosphatase of 245 units/L (normal range 30–115 units/L) is also noted from the liver function tests.

Q11. What form of renal osteodystrophy is likely to have contributed to Mrs A’s fracture and how does it influence bone turnover? On admission to the orthopaedic ward, Mrs A was taking the following renal bone disease drug regimen: • Sevelamer 800 mg three tablets three times a day • Alfacalcidol 0.75 µg once a day. The nephrology team have decided to increase her sevelamer to four tablets three times a day and will consider adding in aluminium hydroxide as an additional phosphate binder if hyper- phosphataemia not controlled in four weeks. They have told Mrs A that they want to look after her bones better and also reduce her ‘calcification risk’.

Q12. Why are the doctors concerned about extraskeletal manifestations of an abnormal bone bio- chemistry profile (i.e. calcification) in Mrs A? At which sites in the body can calcification occur in a CKD patient? How can calcification impact on the morbidity and mortality of CKD patients?

Q13. What are the benefits of prescribing higher doses of calcium-free phosphate binders (sevelamer and/or aluminium hydroxide) for Mrs A in relation to preventing further bone fractures and reducing her calcification risk? 74 Chapter 6 • Renal bone disease

References haemodialysis patients. Am J Kidney Dis 1999: 33: 694–701. 13. Block GA, Sakiewicz PG. Serum bicarbonate levels 1. Sherrard DJ, Hercz G, Pei Y et al. The spectrum of in sevelamer versus calcium containing phos- bone disease in end stage renal failure – an evolv- phate binders in haemodialysis patients. J Am Soc ing disorder. Kidney Int 1993; 43: 436–442. Nephrol 2001; 12: 761A. 2. Malluche H, Faugere MC. Renal bone disease 14. Slatopolsky EA, Burke SK, Dillon MA. Renagel, a 1990: an unmet challenge for the nephrologists. nonabsorbed calcium and aluminium free phos- Kidney Int 1990; 38: 193–211. phate binder, lowers serum phosphorus and 3. Davies MR, Hruska KA. Pathophysiological mech- parathyroid hormone. Kidney Int 1999; 55: anisms of vascular calcification in end stage renal 299–307. disease. Kidney Int 2001; 60: 472–479. 15. Herrmann P, Ritz E, Sckmidt-Gayk H et al. 4. Foley RN, Palfrey PS, Saran MJ. Clinical epidemi- Comparison of intermittent and continuous oral ology of cardiovascular disease in chronic renal administration of calcitriol in dialysis patients: a disease. Am J Kidney Dis 1998; 32: S112–S119. randomised prospective trial. Nephron 1994; 67: 5. Causes of death. In: Renal Data System. USRDS 48–53. 1998 annual data report. Bethesda, MD: National 16. Lee WT, Padayachi F, Collins JF et al. A compar- Institute of Diabetes and Digestive and Kidney ison of oral and intravenous alfacalcidol in the Diseases, 1999: 79–90. treatment of uraemic hyperparathyroidism. J Am 6. Gnash SK, Stack AJ, Levin NW et al. Association Soc Nephrol 1994; 5: 1344–1348. of elevated serum phosphate, calcium phos- 17. Renal Association. Treatment of Adults and Children phate product and parathyroid hormone with with Renal Failure: Standards and Audit Measures, cardiac mortality risk in chronic haemodialysis 3rd edn. London: Lavenham Press, 2002. patients. J Am Soc Nephrol 2001; 12: 2131–2138. 18. National Kidney Foundation. K/DOQI clinical 7. Lowrie EG, Lew NL. Death risk in haemodialysis practice guidelines for bone metabolism and dis- patients: the predictive value of commonly ease in chronic kidney disease. Am J Kidney Dis measured variables and an evaluation of death 2003; 42 (Suppl 3): S1–S201. rate differences between facilities. Am J Kidney Dis 19. Hamdy NAT, Kanis JA, Beneton MNC et al. Effect 1990; 15: 458–482. of alfacalcidol on natural course of renal bone 8. Block GA, Hulbert-Sheron TE, Levin NW, Port FK. disease in mild to moderate renal failure. BMJ Association of serum phosphate and calcium 1995; 310: 358–363. phosphate product with mortality risk in chronic 20. Raggi P, Chertow GM, Bommer J et al. Cardiac haemodialysis patients: a national study. Am J calcification is prevalent and severe in ESRD Kidney Dis 1998: 31; 607–617. patients as measured by electron beam CT scan- 9. Young E, Satayathum S, Pisoni RL et al. ning. J Am Soc Nephrol 2000; 11: 75A. Prevalence of values on mineral metabolism 21. Goodman WG, Golden J, Kuizon BD et al. being outside the targets from the proposed new Coronary artery calcification in young adults draft NKF-K/DOQI and European Best Practice with end stage renal disease who are undergoing Guidelines in countries of the dialysis outcomes dialysis. N Engl J Med 2000; 342: 1478–1483. and practice patterns study (DOPPS). Nephrol Dial 22. Chertow GW, Burke SK, Raggi P. Sevelamer at- Transplant 2003; 18 (Suppl 4): 677–678. tenuates the progression of coronary and aortic 10. Janssen MJA, van de Kuy A, ter Wee PM, van calcification in haemodialysis patients. Kidney Int Boven WPL. Aluminium hydroxide, calcium 2002; 62: 245–252. carbonate and calcium acetate in chronic inter- 23. Drueke TD. Control of secondary hyperpara- mittent haemodialysis patients. Clin Nephrol thyroidism by vitamin D derivatives. Am J Kidney 1996; 45: 111–119. Dis 2001; 37: S58–S61. 11. Altmann P. Aluminium toxicity in dialysis patients: no evidence for a threshold serum alu- minium concentration. Nephrol Dial Transplant 1993; 8: 25–34. Further reading 12. Bleyer AJ, Burke SK, Dillon MA et al. A compar- ison of the calcium free phosphate binder sevelamer hydrochloride with calcium acetate Altmann P. Calcium and phosphate in renal failure: in the treatment of hyperphosphataemia in the disease. Br J Renal Med 2001; Winter; 6–9. Further reading 75

Altmann P. The control of calcium and phosphate in chronic kidney disease. Am J Kidney Dis 2004; 43: renal failure. Br J Renal Med 2002; Spring: 6–9. 572–579. Block GA, Friedrich K. Re-evaluation of risks associ- Hruska KA, Teitelbaum SL. Renal osteodystrophy. N ated with hyperphosphataemia and hyperpara- Engl J Med 1995; 333: 166–174. thyroidism in dialysis patients: recommendations Hudson JQ. Improved strategies for the treatment of for a change in management. Am J Kidney Dis renal osteodystrophy. J Pharm Pract 2002; 15: 2000; 35: 1226–1237. 456–471. Goodman WG, London G. Vascular calcification in

7

Hypertension and hyperlipidaemia

Clare Morlidge

The aim of this chapter is to cover the basics example a 5 mmHg increase in diastolic BP about hypertension and hyperlipidaemia in results in a 35–40% increase in risk of stroke.1 A general, and with respect to a renal patient. 5 mmHg decrease in BP is associated with a Within hypertension, assessment and dia- 25% reduction in risk of renal failure.1 The gnosis of the hypertensive patient will be absolute benefits of lowering BP depend on covered, followed by the treatment, both the underlying level of risk in an individual pharmacological and non-pharmacological. The subject. The majority of end stage renal failure principles for management of hypertension in (ESRF) patients will die from a cardiovascular the non-renal patient apply to the renal patient complication. too, but with a few exceptions, which will be The kidneys’ main function is to remove covered. waste products, toxins and excess fluid from the Within hyperlipidaemia, the causes of dys- body. One of their extra functions is in the con- lipidaemia are examined followed by a risk trol of BP. They achieve this by the renin– assessment and treatment options. angiotensin aldosterone cascade, which is important for longer term salt, water and BP control. A high BP can be the cause of renal impairment or a complication of it. It can be Hypertension difficult to determine whether the high BP has caused the renal impairment or whether the Blood pressure distribution within the popula- renal impairment has caused the high BP. tion displays a bell-shaped curve and there is no The majority of chronic renal failure patients clear cut-off point between hypertensive and are affected by hypertension, and ischaemic normotensive subjects. There is, however, con- heart disease (IHD) has higher prevalence than siderable evidence that treatment of subjects in the general population. Both hypertension with a blood pressure (BP) above the thresholds and chronic fluid overload can lead to left currently used (see below), results in important ventricular hypertrophy (LVH), which in turn clinical benefits. results in left ventricular failure (LVF). LVH has The complications of hypertension include: been shown to be a predictor of mortality in haemodialysis patients. • Stroke • Myocardial infarction • Other end organ damage (e.g. kidneys) What is normal blood pressure? • Malignant hypertension • Peripheral vascular disease. Both systolic and diastolic BPs increase with age, The most common of these are myocardial so there is a normal range for each age (Table infarction (MI) and stroke. Small reductions in 7.1).2 The British Hypertension Society guide- BP result in substantial reductions in the risk of lines (2004) state that target BP for diabetics is developing a cardiovascular complication. For 130/80 and for non-diabetics is 140/85.3

77 78 Chapter 7 • Hypertension and hyperlipidaemia

wrongly diagnosed with hypertension and may Table 7.1 Normal range of blood pressure at receive unnecessary treatment with drugs. To different ages take a reading the arm should be supported level with the heart. BP is measured by listening Age Blood pressure (mmHg) for the Korokoff sounds. Read systolic BP Systolic Diastolic when sounds first appear and diastolic BP when sounds disappear. <30 years 100–120 60–70 The BP should be measured several times 30–60 years 110–130 70–80 over several visits in order to diagnose hyper- >60 years 120–140 80–90 tension. If the first reading is dangerously high, however, several readings should be taken on Data taken from Kidney Failure Explained.2 the first visit.

Causes of hypertension Assessment of the hypertensive patient In 90–95% of cases of hypertension there is no The patient should be assessed to detect any end underlying medical illness to cause the organ damage, for example, the optic fundi increased blood pressure. The remaining 5–10% should be inspected for damage to the eyes, of cases are secondary to another disease urinalysis and urea and electrolytes (U&Es) process, such as renal disease, endocrine dis- carried out for the kidneys, and cholesterol levels eases (e.g. Conn’s syndrome), vascular disease should be checked. An electrocardiogram (ECG) (e.g. renal artery stenosis) or drugs (e.g. oestro- should be done to check for ventricular hyper- gens, ciclosporin, erythropoietin). trophy, and echocardiography (ECHO) should be performed if ventricular hypertrophy is sus- Clinical presentation pected after ECG. Abdominal ultrasound should be performed if renal impairment is suspected. Hypertension itself causes no symptoms, some The patient’s history should be taken in full people with high BP suffer from a headache, but to determine if there is a secondary cause for the this is not reliable. Often there are no symptoms increased blood pressure. Even in renal patients until end organ damage occurs (e.g. MI or a careful history should be taken and all the stroke). tests performed to help to determine the cause Hypertension usually comes to light at a rou- of hypertension. This helps to determine if the tine health check or following a hypertensive- renal impairment is the cause of hypertension related complication (e.g. stroke). or a consequence of it. There may be a second cause of hypertension. The patient should be assessed for contribut- Diagnosis ing factors (e.g. obesity, increased cholesterol). Lifestyle should be addressed and changes made Blood pressure is measured using a sphygmoma- if appropriate. Patients should be advised to: nometer (sphyg). Two readings are taken. The • Avoid smoking first shows the systolic BP (when the heart con- • Exercise regularly tracts), and the second shows the diastolic BP • Reduce alcohol intake (when the heart relaxes), giving a reading of, for • Reduce salt intake example, 140/80. An increase in both systolic • Avoid being overweight and diastolic BPs is a risk factor for stroke and • Stick to fluid restriction if appropriate. coronary heart disease (CHD). It is important to use a correctly sized cuff, The risks of stroke (sixfold greater in hyper- since one too small will overestimate the tensive patients than in normotensive patients), patient’s BP. As a result, a person may be coronary heart disease (threefold increase) and Hypertension 79 peripheral vascular disease (twofold increase) The aims of BP management are to: are multifactorial.1 Other risk factors (e.g. smok- • Attain good BP control ing, diabetes) act as multipliers determining • Reduce cardiovascular morbidity and individual risk. Ischaemic heart disease (IHD) mortality has a much higher prevalence in the renal pop- • Prevent atherosclerosis ulation than in the general population. • Control other vascular risk factors • Reverse end organ damage. Self-help measures These are all important in the renal patient as well as the non-renal patient. In renal • Avoid being overweight – There is a strong patients, especially those on dialysis, chronic link between being overweight and having fluid overload is treated by fluid removal and hypertension. Patients whose weight is above optimisation of the patient’s dry weight. that advised for their height should be Patients are advised to stick to their recom- advised to lose weight. Even if not over- mended fluid intake and to restrict the sodium weight, patients should be advised to follow in their diet. High-dose loop diuretics help with a low-fat, high-fibre diet. Refer to a dietician fluid overload. if appropriate. • Keep alcohol levels down – There is a strong link between a high alcohol intake and raised Drug treatment BP. Drinking a moderate amount is harmless and may even be beneficial for coronary Drugs commonly used in the treatment of heart disease (CHD). However excessive hypertension are: alcohol consumption can cause a resistance to antihypertensive drugs. • Thiazides • Reduce salt intake – The relationship • Beta-blockers between salt intake and BP is difficult to • Alpha-adrenoceptor blockers prove, but there is good evidence to suggest • Calcium channel blockers that a high salt diet affects your BP. Salt can • Angiotensin-converting enzyme (ACE) also increase the amount of fluid that your inhibitors body retains. Restricting salt intake to • Angiotensin II receptor blockers 100 mmol (6 g) per day combined with • Centrally acting drugs weight reduction does reduce cardiovascular • Direct acting vasodilators. risk in patients with hypertension and this is Tight BP control, especially in renal patients, advised in many renal patients. Salt is hidden can be difficult to obtain, and may result in in many processed foods. Advise patients to patients being on maximum doses of many eat mainly fresh, unprocessed foods. antihypertensive drugs. In renal patients drugs • Exercise regularly – Exercise can help to should be started at low doses and increased reduce your BP and keep weight down. It is cautiously. Refer to the British National also a good stress reliever. Aim for 20–30 Formulary or individual summary of product minutes of brisk activity at least three times characteristics (SPCs) for dosing information a week (e.g. brisk walk, swimming, etc.). and to the Renal Drug Handbook4 for dosing in • Stop smoking – Giving up smoking does not renal impairment. Beta-blockers, calcium chan- improve BP control, but it does lower the risk nel blockers and ACE inhibitors have all been of blood vessel damage that can lead to an shown to improve LVH in the general popula- MI or stroke. tion and are safe in chronic renal failure. These non-pharmacological interventions The British Hypertension Society recom- can bring down BP a little, but more import- mendations are shown in Figure 7.1.3 antly can reduce the overall risk of end organ The ASCOT trial recommends using ACE damage. inhibitors first line for younger patients and 80 Chapter 7 • Hypertension and hyperlipidaemia

Younger (< 55 years) Older (> 55 years) and non-black or black

Step 1 A or (B*) C or D

Step 2 A or (B*) + C or D

Step 3 A or (B*) + C + D

Step 4 Step 3 + E Resistant hypertension

Figure 7.1 British Hypertension Society recommendations for combining blood pressure-lowering drugs. A = angiotensin-converting enzyme (ACE) inhibitor or angiotensin II receptor blocker; B = beta-blocker; C = calcium channel blocker; D = thiazide diuretic; E = add either alpha-blocker or spironolactone or other diuretic. *Combination therapy involving B + D may induce more new onset diabetes compared with other combination therapies. Adapted from ref. 3. calcium channel blockers for older patients.5 system event, in particular stroke. Beta-blockers The National Institute for Health and Clinical are an alternative to an ACE inhibitor when an Excellence (NICE) recommends: ACE inhibitor is contraindicated or not toler- ated (e.g. women of childbearing age or those • For patients over 55 years old or black – a cal- with an increased sympathetic drive).6 cium channel blocker or thiazide diuretic In patients with renal disease, hypertension first line, ACE inhibitor second line, and a can increase the rate of progression to end stage calcium channel blocker or thiazide third renal disease (ESRD) and therefore the use of line whichever was not used first. drugs can be important in delaying the progres- • For patients under 55 years old, an ACE sion to ESRD. This is the same for all causes of inhibitor first line, a calcium channel blocker renal impairment. In some cases initiation of or thiazide second line and a calcium chan- dialysis is needed to control hypertension. In nel blocker or thiazide third line. diabetics and some other renal diseases (e.g. • For all patients an alpha-blocker, further glomerulonephritis), controlling the BP with an diuretic or beta-blocker as a fourth drug with ACE inhibitor or angiotensin II blocker slows referral to a specialist. the damage to the kidney and therefore delays NICE also recommends that beta-blockers are the time to ESRD. ACE inhibitors should be not used as an initial therapy for hypertension used with caution in other patients with renal since they have been shown to be less effective disease since they can increase the damage to at reducing the risk of a major cardiovascular the kidney and therefore reduce the time until Hyperlipidaemia 81 dialysis is required. In all patients, when start- to the liver where they are cleared from circula- ing an ACE inhibitor or angiotensin II blocker, tion. renal function should be measured prior to High-density lipoprotein (HDL) transports starting therapy and regularly thereafter to pick cholesterol from peripheral tissues to the liver, up if the drug is affecting the patient’s renal and plays a major role in maintaining cho- function. lesterol homeostasis. About 30% of blood In renal artery stenosis (RAS) the main blood cholesterol is carried by HDL. In turn, low- vessel running to the kidney becomes blocked – density lipoprotein (LDL) transports cholesterol partially or fully – due to atherosclerosis. It from the liver via the plasma to those cells that usually affects both kidneys. It is a common require it. Cholesterol is an essential part of cell cause of renal failure in older people, especially membranes and is a precursor of steroid hor- those who have suffered an MI or stroke (i.e. mones. LDL is the main lipoprotein involved in are at increased risk of atherosclerosis). In atherosclerosis.1 patients with RAS, ACE inhibitors and Elevated concentrations of total cholesterol angiotensin II blockers are generally avoided (TC) and LDLs increase the risk of CHD, while since they can wipe out renal function com- HDLs confer protection. The term dyslipidaemia pletely, accelerating the progression to ESRD. is a more appropriate term to use when consid- Occasionally, if the benefits outweigh the risks ering the risk of CHD. these drugs may be used under the close super- An ideal serum lipid profile would be:1 vision of a nephrologist.7 • TC <5.0 mmol/L Thiazide diuretics are ineffective once the • LDL <3.0 mmol/L glomerular filtration rate (GFR) becomes less • Triglycerides <2.3 mmol/L than 25 mL/min, and loop diuretics are often • HDL >0.9 mmol/L. used at high doses (e.g. furosemide 500 mg to 1 g daily) to gain an effect. Metolazone is effec- tive when combined with a loop diuretic. Potassium-sparing diuretics such as amiloride Primary dyslipidaemia are not recommended. Spironolactone is not generally used, but is beneficial in low dose for This can be genetically determined although the treatment of heart failure even in patients expression is influenced by environmental fac- on dialysis. Beta-blockers and calcium channel tors. Familial hypercholesterolaemia is the most blockers are generally well tolerated. Any ankle common inherited dyslipidaemia. It is caused swelling with calcium channel blockers must by a mutation in the LDL receptor gene. not be confused with fluid overload. Patients with the genetic dyslipidaemia will be Moxonidine, although contraindicated in severe at increased risk of MI, or stroke, due to elevated renal impairment, is used in practice. concentrations of cholesterol and triglycerides.

Secondary dyslipidaemia Hyperlipidaemia Dyslipidaemia can be secondary to diet, drugs or a number of other disorders. These dyslipi- When dietary cholesterol and triglycerides are daemias are more easily corrected than primary absorbed from the intestine they are transported dyslipidaemia. Common causes of secondary in the intestinal lymphatics as chylomicrons. dyslipidaemia include: These pass through blood capillaries where the enzyme lipoprotein lipase catalyses the break- • Diabetes mellitis down of triglycerides to free fatty acids and gly- • Chronic renal failure cerol, which enter adipose tissue and muscle. • Nephrotic syndrome The cholesterol-rich chylomicron remnants go • Drugs. 82 Chapter 7 • Hypertension and hyperlipidaemia

Diabetes mining a higher risk of progression to dialysis. Statin therapy has been shown to prevent creat- The incidence of CHD is up to four times higher inine clearance decline and to slow renal func- in diabetic patients than in the general popula- tion loss, particularly in case of proteinuria and tion. Many renal patients have diabetes so this this effect may only partially be due to their is a risk factor for them. In people with diabetes favourable effect on hyperlipidaemia.11,12 any atherosclerotic disease is often more wide- spread, and plaque rupture and thrombotic occlusion occur more often. It is the main cause Drugs of reduced life expectancy in people with dia- Many drugs can adversely affect the lipid pro- betes. The results of the Heart Protection Study file. Hypertension is a major risk factor for suggest that statin therapy reduces vascular atherosclerosis and there is concern that whilst events regardless of cholesterol level.8 treating hypertension has reduced the incidence of MI, stroke and renal failure, it has had no Chronic renal failure major impact on reducing CHD.

Dyslipidaemia is common in patients with Antihypertensive drugs impaired renal function. The hypertriglyceri- Many of the antihypertensive drugs themselves daemia is associated with reduced lipoprotein have an adverse effect on lipids. Thiazide diuret- lipase activity. In chronic renal failure, diabetes, ics and beta-blockers are the main culprits. The type of renal disease, transplantation and drugs benefits of these drugs, for example following 9 can all play a part in the dyslipidaemia (see also MI or in heart failure, outweigh any benefit of Chapter 9). Dyslipidaemia often accelerates withdrawal. However other antihypertensive renal disease partly by promoting renal fibrosis drugs (e.g. ACE inhibitors or calcium channel in early atherosclerosis. The pathways for this blockers) are without adverse effects on lipids fibrosis may be reversible and early intervention and these could be used as alternatives in those 10 could preserve the kidney. patients more at risk.

Ciclosporin Nephrotic syndrome Ciclosporin has been associated with increased This is a syndrome in which the kidney’s LDL. Its use is widespread in renal patients nephrons becomes ‘leaky’ and allow protein to either following transplantation or in certain enter into the urine. There are many causes of diseases (e.g. nephrotic syndrome). This adverse nephrotic syndrome, and a biopsy will deter- effect is often compounded by the concomitant mine the cause. These patients have an increase use of prednisolone. This combination con- in circulating lipoproteins, which is related to tributes to the adverse lipid profile seen follow- the extent of proteinuria and serum albumin ing renal transplantation (see Chapter 9). Statins level. HDL levels are usually unchanged. The improve the profiles of those atherogenic lipids use of steroids may exacerbate the underlying associated with the hypercholesterolaemia seen lipoprotein abnormality. Many of these patients in renal transplant patients treated with the require treatment with statins, but intervention immunosuppressants ciclosporin or tacrolimus.9 is often delayed until after a trial of steroids to The combination of statins with ciclosporin see if the disease is controlled, since as the level increases the risk of myositis and rhabdomyo- of proteinuria falls and serum albumin increases lysis and therefore should be used with caution the cholesterol level may return to normal. If (its use is widespread). biopsy shows the nephrotic syndrome is not autoimmune or steroid responsive, then statins Sirolimus may be started immediately. Hyperlipidaemia Sirolimus is associated with a marked hyper- itself can affect renal function, increase protein- lipidaemia, including hypercholesterolaemia uria and speed glomerulosclerosis, thus deter- and hypertriglyceridaemia. Cholesterol levels Hyperlipidaemia 83 should be measured prior to starting treatment, part (e.g. plaque stabilisation, inhibition of and regularly thereafter to monitor extent of thrombus formation and anti-inflammatory hyperlipidaemia, and treatment initiated if activity).1,10,11 All statins have the potential to necessary.13 cause muscle myopathy and this is more pre- valent in renal impairment and with concom- itant use of ciclosporin.8 This effect is also seen Risk assessment with tacrolimus and sirolimus. Therefore all statins should be started at low doses and 14 The Sheffield table identifies patients who increased with caution in patients with renal should have their lipid levels measured, and impairment.4 those who would benefit from treatment. The threshold for commencing treatment is a risk of developing CHD of 30% over 10 years. Fibrates Patients with CHD and levels of TC >5 mmol/L Fibrates reduce triglycerides and LDL whilst and LDL >3 mmol/L are most likely to benefit increasing HDL. They also have a beneficial from treatment.14 effect on fibrinolytic and clotting mechanisms. Myositis is also associated with fibrates, espe- Treatment cially in renal impairment or in combination with statins, and as with the statins doses need Before starting drug therapy other risk factors to be adjusted with respect to renal function.4 should be tackled, such as obesity, smoking, high alcohol intake and lack of exercise. Ezetimibe Underlying disorders, such as hypertension, should be treated. A low-fat diet should be com- Ezetimibe inhibits the absorption of cholesterol menced. The patient should be informed that from the intestine. It can be used either alone treatment involves a long-term commitment to or in combination with a statin. It has been well drug therapy and also appropriate dietary and tolerated, with no dosage adjustment required lifestyle changes. in renal impairment.4 However, it can affect The two most widely used drugs are statins ciclosporin levels, and ciclosporin can increase and fibrates. Doses may need to be adjusted ezetimibe levels, so the combination should be according to renal function.15 used with caution and levels monitored carefully.16 Statins In patients with CHD or other occlusive arterial Statins inhibit the enzyme 3-hydroxy-3-methyl- disease (e.g. RAS), treatment should include: glutaryl coenzyme A reductase (HMG-CoA). Their primary site of action is in the liver, where • Statin to lower TC <5 mmol/L they inhibit the rate-limiting step in the bio- • Advice to stop smoking synthesis of cholesterol. The effect of statins on • Tight control of BP to <140/90 lipid profile contributes to their beneficial out- • Information on risk factors (e.g. diet, alcohol, come in reducing morbidity and mortality in weight) CHD. Other mechanisms may also play a • Low-dose aspirin (75 mg daily). 84 Chapter 7 • Hypertension and hyperlipidaemia

C CASE STUDY

Mr BJ is a 50-year-old man whose blood pressure has been consistently above 140/85 for six months although he has not been started on any antihypertensive therapy. He is a type 2 diabetic but has no history of coronary heart disease. On a routine clinic visit he is found to have a blood pressure of 150/95, a creatinine of 160 µmol/L a total cholesterol of 5.5 mmol/L and HDL 0.8 mmol/L.

Q1. What risk factors does he have for CHD?

Q2. What treatment would be suggested with respect to his blood pressure?

Q3. What other advice would be appropriate?

References 9. Imamura R, Ichimaru N, Moriyama T et al. Long term efficacy of simvastatin in renal transplant recipients treated with ciclosporin or tacrolimus. 1. Walker R, Edwards C. Clinical Pharmacy and Clin Transplant 2005; 19: 616–621. Therapeutics, 3rd edn. Edinburgh: Churchill 10. Chade AR, Mushin OP, Zhu X et al. Pathways Livingstone, 2003. of renal fibrosis and modulation of matrix 2. Stein A, Wild J. Kidney Failure Explained, 2nd edn. turnover in experimental hypercholesterolemia. London: Class Publishing, 2002. Hypertension 2005; 46: 772–779. 3. Williams B, Poulter NR, Brown MJ et al. The BHS 11. Buemi M, Nostro L, Crasci et al. Statins in Guidelines Working Party. British Hypertension nephrotic syndrome: a new weapon against tis- Society Guidelines for Hypertension Manage- sue injury. Med Res Rev 2005; 25: 587–609. ment, 2004 – BHS IV: Summary. BMJ 2004; 328: 12. Kronenberg F. Dyslipidemia and nephritic syn- 634–640. drome: recent advances. J Ren Nutr 2005; 15: 4. Ashley C, Currie A. The Renal Drug Handbook, 2nd 195–203. edn. Oxford: Radcliffe Medical Press, 2004. 13. Wyeth. Summary of product characteristics for 5. Dahlof B, Sever PS, Poulter NR et al. for the Sirolimus. January 2005. http://emc.medicines. ASCOT investigators. Prevention of cardiovascu- org.uk lar events with an antihypertensive regimen, in 14. Wallis EJ, Ramsay LE, Haq IU et al. Coronary and the Anglo-Scandinavian Cardiac Outcomes Trial cardiovascular risk estimation for primary pre- (ASCOT). Lancet 2005; 366: 895–906. vention: validation of a new Sheffield table in the 6. National Institute for Health and Clinical 1995 Scottish Health Survey population. BMJ Excellence (NICE). Hypertension: Management of 2000; 320: 671–676. Hypertension in Adults in Primary Care. Clinical 15. Joint Formulary Committee. British National Guideline CG34. NICE, June 2006 (accessed from Formulary. 53 edn. London: British Medical www.nice.org.uk). Association and Royal Pharmaceutical Society of 7. Stein A, Wild J. Kidney Dialysis and Transplants. Great Britain, 2007. London: Class Publishing, 2002. 16. Kosoglou T, Stalkevich P, Johnson-Leanos AO et 8. Heart Protection Study. MRC/BHF Heart al. Ezetimibe: a review of its metabolism, phar- Protection Study of cholesterol lowering with macokinetics and drug interactions. Clin simvastatin in 20536 high risk individuals: a ran- Pharmacokinetics 2005; 44: 467–494. domised placebo-controlled trial. Lancet 2002; 360: 7–22. 8

Renal replacement therapy

Aileen Currie and James Dunleavy

In renal failure the normal function of the permeable membranes and diffusion to separate kidney deteriorates with time and may be lost colloids and crystalloids. He termed this process completely. In the initial ‘pre-dialysis’ phase the ‘dialysis’. Problems in developing suitable vas- use of drugs, modification of diet and fluid cular access and anticoagulation, however, restriction can delay the patient’s progression to delayed the introduction of dialysis as a useful stage 5 chronic kidney disease (CKD stage 5), treatment. The first successful human dialysis where the glomerular filtration rate (GFR) was carried out in Germany in 1924 by George <15 mL/min. However the resultant loss of Haas. Willem J Kolff then developed the first homeostatic function results in an accumula- dialyser in the Netherlands and used it as a tion of waste products, fluid retention and treatment for acute renal failure in 1943. abnormal electrolyte levels. Therefore most Despite its long history, dialysis has only been patients with severe chronic renal failure will available as a treatment for chronic renal disease eventually require some form of renal replace- since the 1960s.1 ment therapy, the primary aim being to correct Although haemodialysis is mainly a hospital- the accumulation of toxins, electrolytes and based treatment it can also be performed at fluid. In acute renal failure it is also used as a home, in a nurse-led satellite unit or even on short-term measure to ‘rest’ the kidneys in the holiday in a unit abroad or in the UK. The hope they will recover function. These patients majority of patients in the UK are dialysed in may be on it for anything from 1 day to 1 year. hospitals, usually three times a week, although The five main treatment options for CKD there is interest at the moment in daily dialysis, stage 5 are: which has been shown to improve adequacy, hypertension and patient quality of life. • Haemodialysis Unfortunately there are financial constraints to • Continuous renal replacement therapy, this at the moment; most units do not have including CVVH, CAVH, CVVHD, CAVHD, space to dialyse patients every day.2 CVVHDF, CAVHDF Haemodialysis works by a combination of • Haemodiafiltration diffusion (the movement of solutes from fluid • Peritoneal dialysis with a high to a low concentration across a • Transplantation. semi-permeable membrane) and ultrafiltration The first four will be discussed in this chapter (the movement of fluid under pressure across a and transplantation is covered in Chapter 9. semi-permeable membrane) (Figure 8.1). Excess fluid is removed by ultrafiltration and waste products by diffusion. Essential minerals (e.g. calcium and bicarbonate) are also replaced by Haemodialysis diffusion.3

A Scottish chemist, Thomas Graham, in the 1850s, first discovered the capacity of semi-

85 86 Chapter 8 • Renal replacement therapy

Physical basis of dialysis Semipermeable membrane

Erythrocyte, red blood cell Bacteria

Albumin, as example of a big protein molecule Medium sized molecules, e.g. β2-microglobulin

Electrolytes Water flow is easily possible

The semipermeable membrane functions similar to a fine sieve, only molecules that are small enough can pass.

Figure 8.1 Diffusion process in relation to dialysis. Copyright and courtesy of Fresenius Medical Care.

Vascular access either be created at the wrist (radial artery) or the elbow (brachial artery). Although brachial There are three methods of vascular access for fistulas have a success rate of 90% with only haemodialysis: 60% for radial fistulas, the latter are usually • Fistula attempted first as once a patient has had a • Catheter brachial fistula they are unable to have a radial • Graft. one. It takes about 6–8 weeks in order for the fistula to be ready for use and therefore it is best to be planned in advance, but unfortunately Fistula this does not always happen.6 They last longer A fistula is usually created as a day surgery case than central venous catheters and 60–90% are under either local or general anaesthesia usually still functioning after 3 years. The depending on the patient’s medical condition. patient must also have a good vascular system It is the surgical connection of an artery to a but 30% of patients do not have suitable veins vein by direct surgical anastomosis; some (e.g. elderly patients or patients with diabetes)2 arterial blood will be diverted due to pressure (Figure 8.2). differences into the vein causing the wall to The main complications are thrombosis, thicken and the lumen to get bigger. This allows which may be due to hypotension, dehydration blood at arterial pressure into the venous system or prolonged compression of the fistula (either close to the surface of the skin so the nurses or accidentally or on purpose) and steal syndrome, the patient are able to get needles into the vessel where too much blood is drawn away from the easily. Fistula formation was first introduced in hand and the fistula usually requires to be tied 1966 and is the preferred option for vascular off (more common with brachial fistulas). It is access for haemodialysis.4 The Renal Association very important that medical and nursing staff standards state that 80% of people should be do not take blood from or put venflons into any dialysed via a fistula (the Scottish Renal veins which have been used or may later be Association want at least 70%).5 Fistulas can used for fistulas.2,7 Haemodialysis 87

Cephalic vein Radial artery

Site of fistula

Figure 8.2 Arterio-venous fistula. Copyright and courtesy of Fresenius Medical Care.

Catheter be either dual or triple lumen. The main prob- lems with catheters are thrombosis and infec- Temporary or permanent central venous tions. Lines may be kept patent by locking them catheters are usually placed into the internal with sodium chloride 0.9% or heparin after use. jugular vein. They can also be placed in the If they become blocked (usually due to fibrin femoral vein if there are problems with the in- sheath formation) then locks or infusions of ternal jugular vein. The main problem with the urokinase or alteplase may have to be used. femoral vein is the high risk of infections and Low-dose warfarin therapy may be initiated if it they tend to only be kept in for short periods of becomes a chronic problem, aiming for an INR time. The subclavian vein is generally not used of 1.5–2.5.3,4,7 because of the risk of venous stenosis.4 Permanent catheters are inserted in radiology or theatre and are tunnelled and have Dacron Graft cuffs to keep infection at bay and keep them in place. Temporary lines are usually inserted at Polytetrafluoroethylene (PTFE) grafts are usually ward level if dialysis is only required for a few only used in the UK if access is a problem. A weeks or until a slot for a permanent catheter plastic tube connects usually the basilic vein to can be organised (ideally they should not be the brachial or radial artery, although they can kept in for more than three weeks). They may also be placed in the thigh. They have the 88 Chapter 8 • Renal replacement therapy advantage that they can be used within 2–4 membrane in the dialyser is reduced.3 The weeks or if required almost immediately after machines are heat sterilised after every patient, placement. The disadvantages are that they chemically sterilised twice a week and heat dis- have an increased infection risk, thrombosis, infected and decalcified every second day. they are difficult to remove and usually only The semi-permeable membrane used for last for 3–5 years.2,7 haemodialysis is known as the dialyser or filter or kidney. This is the major high-cost item in haemodialysis. It consists of thousands of long Process hollow fibres enclosed in a rigid polyurethane shell. There are two ports on each side to allow For haemodialysis, blood is removed from the the blood and dialysate to flow in and out. The patient from the arterial site via either the line or blood flows through the fibres and the dialysate fistula and pumped through the dialysis machine surrounds the fibres. The blood and dialysate via the dialyser where diffusion and ultrafiltra- flow in opposite directions, i.e. countercurrent, tion occur. The blood is then returned to the allowing diffusion and ultrafiltration to take patient via the venous site. The blood and place more effectively. The dialyser allows the dialysate flow in opposite directions to optimise free movement of low-molecular-weight the removal of waste products. Pumps are needed molecules (<5 kDa) but restricts the passage of to produce a high enough flow rate for the blood larger molecules and blood.3 and dialysate to go round the extracorporeal cir- cuit and to control the ultrafiltration rate, blood pressure and dialysate pressure (Figure 8.3). The Types of dialyser blood flow rate is ideally 200–500 mL/min and There are many different dialysers available, the the dialysate flow rate 500 mL/min, which can be main differences are:2,7,9,10 increased to 800 mL/min, although there is not a huge increase in clearance with the latter. As • Shape: Hollow fibre (capillaries) – this is the dialysis progresses, the efficiency is decreased most common form – or flat plate (parallel because the concentration gradient across the plates).

Haemodialysis Flow scheme haemodialysis

Dialyser Anticoagulation Blood pump

Blood to the patient

Fresh dialysate

Blood from the patient Used dialysate

Figure 8.3 Haemodialysis circuit. Copyright and courtesy of Fresenius Medical Care. Haemodialysis 89

␤ • Surface area: The bigger the surface area the medium-sized molecules (e.g. 2-microglobulin) ␤ more dialysis the patient receives. A typical as small molecules. Accumulation of 2- dialyser has a surface area of 1–2 m2. microglobulin can lead to problems in the long • Method of sterilisation: Steam, ethylene term with amyloidosis that can present as carpal oxide (this must be rinsed out before use due tunnel syndrome, arthropathy and bone to its toxicity), gamma-irradiation. cysts.9,11 • Material: Cellulose (can cause complement An interesting side-effect occurs between and leukocyte activation) (e.g. cuprophan), ACE inhibitors and high-flux polyacrylonitrile modified cellulose (e.g. cellulose acetate, membranes, where anaphylaxis can occur due ␤ haemophan), synthetic (best for 2- to bradykinin formation between the dialyser microglobulin removal) (e.g. polysulfone, and the patient’s blood. This type of dialyser is polyamide, polyacrylonitrile). no longer routinely used for haemodialysis.11,14 • Membrane permeability: The more per- meable the dialyser is to fluid the higher its flux, measured as ultrafiltration coefficient Dialysate

(Kuf) and varies from 2 to 85 mL/h/mmHg. Low flux has a Kuf of 2–10 mL/h/mmHg and The dialysis fluid known as dialysate consists of high flux has a Kuf greater than 20 to over a concentrated solution of electrolytes dissolved 60 mL/h/mmHg. in water. They differ only in respect of the

• Clearance: Of urea, creatinine, vitamin B12, quantities of electrolytes in each (e.g. low or phosphate, inulin and sieving coefficients for high calcium or potassium concentrations), and ␤ albumin (should be 0) and 2-microglobulin. which one is chosen depends on the patient’s requirements. Some now also come with glu- The most biocompatible dialyser is one that cose, which helps prevent hypoglycaemia dur- has been steam sterilised and is made of a ing dialysis in diabetic patients and can reduce synthetic material (e.g. polysulfone). The more disequilibrium syndrome by maintaining the biocompatible the dialyser the fewer long- osmotic pressure. The majority of units will term side-effects a patient is likely to suffer, limit the number of different fluids they use; for although it has not been determined whether example, they may keep a low and a high potas- the short-term complications (e.g. hypotension, sium and calcium fluid. The fluid is brought to headaches and muscle cramps) are related to body temperature by mixing with purified bioincompatibility or rate of fluid and urea water. Bicarbonate is supplied in dry powder removal. Biocompatible membranes are most form as it is too unstable to be mixed with the important for people who may be on dialysis dialysate and the machine calculates the pro- long term.9–11 portion of bicarbonate required.2 Hollow-fibre dialyers can be reused. Initially, The water used to dilute the dialysate must this was to save money and to make the dialyser be purer than drinking water as patients are more biocompatible, although they lose their exposed to approximately 300–400 L of water efficiency the more they are reused. In theory, each week.15 Before entering the dialysis they can be reused up to a maximum of 4–6 machine the water passes through a reverse times, but none are actually licensed for this in osmosis system to remove the aluminium and the UK. There are costs involved in reuse, as biomaterials (e.g. bacteria and other potential specialised machinery and personnel are endotoxins) and this is monitored regularly. required, as well as storage space where the dia- Aluminium enters the water supply as part of lysers are kept labelled for each individual the water treatment process and has been patient. Very few, if any units, reuse them shown to accumulate in people with renal fail- these days due to an increase in morbidity, ure. Signs of aluminium toxicity include reduction in costs of new dialysers and dementia and bone disease. Aluminium levels improved biocompatibility.11–13 can be high in patients who take aluminium Haemodialysis is not as good at removing hydroxide (AluCap capsules) as a phosphate 90 Chapter 8 • Renal replacement therapy binder, although they are rarely used now, and to their body weight without excess fluid or when they are it is only for a short period of causing hypotension. Patients are advised to time. The water supplier limits the aluminium, limit their fluid intake between dialysis sessions calcium, chloramines, nitrates, sodium, sulfates, to as little as 750–1000 mL per day, depending zinc and copper.2 on their urine output. This can be problematic as fluid includes soup, custard, gravy, etc. Sometimes if people have problems tolerat- Anticoagulation ing fluid removal on dialysis, ‘ultrafiltration profiling’ or ‘sodium profiling’ may be attempted. This involves taking more fluid off at Anticoagulation is usually necessary to keep the the beginning of dialysis so the body has time blood flowing through the extracorporeal cir- to normalise before dialysis is finished or alter- cuit. Heparin is used to maintain anticoagula- ing the sodium content of the dialysate fluid tion in most patients during dialysis, with the during the dialysis (start with a high sodium dose being dependent on the response of concentration then reduce it as the dialysis pro- the patient. The majority of units will use gresses). It can reduce the incidence of dizziness conventional heparin although occasionally and cramps due to fluid removal but may exac- low-molecular-weight heparin may be used. erbate thirst, which does not help the patient’s Bemiparin, dalteparin, enoxaparin or tinzaparin efforts at fluid restriction.17 are licensed in the UK for this use but can make Inadequate dialysis and poor nutritional the process of dialysis even more expensive. In intake are associated with increased mortality patients with bleeding complications, either a and morbidity. Inadequate dialysis can also play continuous infusion of epoprostenol may be a major role in erythropoietin resistance, result- used or heparin-free dialysis may be considered. ing in worsening of anaemia. Other drugs used in cases of intolerability to There are two main methods of determining heparin or thrombocytopenia are lepirudin, the adequacy of dialysis: argatroban and danaparoid, none of which are licensed in the UK for this indication.2 More • Urea reduction ratio (URR) information can be found in the Renal Drug • Urea kinetic modelling (UKM) (Kt/V). Handbook by Ashley and Currie.16 URR (as the name suggests) is the difference in urea pre and post dialysis expressed as a per- centage. The target URR for adequate dialysis is Time on haemodialysis and adequacy >65%.5 This method is not ideal, as it does not take into account the effect of residual renal When people first start on dialysis they have function or dietary intake. short and frequent sessions as the body has Urea kinetic modeling (UKM) includes the adjusted, or become accustomed to having large more complex calculation of urea removal Kt/V, quantities of waste products in the blood. If where K is urea clearance per minute, both the these are removed too rapidly a ‘disequilibrium’ patient’s and the dialyser’s, t is time on dialysis syndrome occurs which results in headaches, and V is volume of distribution of urea (i.e. the nausea and vomiting, and in severe cases con- volume of body water in which the urea is fusion and convulsions.3 distributed). The calculation also takes into The length of time on haemodialysis is account protein catabolic rate, nutritional status gradually increased to between 3 and 5 hours and residual renal function so is a more accurate three times a week, the length of time depend- determination of dialysis adequacy. The mathe- ing on the patient’s blood results, size and matical equations used may be complex and are residual urine output. best calculated by computer. They can be used to The patient’s weight is measured before and determine how much dialysis a patient requires.4 after dialysis, the post weight being as close to Other factors that can give an indication of their dry weight as possible, which is equivalent adequacy are normalised protein catabolic rate Continuous renal replacement therapies 91

(normalised for weight), acidosis, weight gain Continuous renal replacement therapies and malnutrition. The Renal Association standards state that an equilibrated Kt/V >1.2 equates to a good dialysis Haemofiltration for three times a week dialysis. If dialysis is only done twice a week then a Kt/V >1.8 is required.5 This method of continuous renal replacement Dialysis adequacy can be increased by:2,11 therapy (CRRT) works by convection (movement of solutes in fluid across a membrane under pres- • Higher blood flow rate sure to remove small, medium and large mole- • Increasing the dialysate flow rate (does not cules) and ultrafiltration (movement of water make a huge difference) across a membrane), which removes extracellular • Increasing the size/surface area of the dialyser fluid with toxins at the filter and adds haemofil- • Longer time on dialysis tration fluid to the filtered blood either before • Changing from haemodialysis to haemo- (pre-dilution) or after (post-dilution) the haemo- diafiltration. filter, so diluting the waste products (Figure 8.4). The common complications associated with The ultrafiltration rate can be controlled by pres- haemodialysis are listed in Table 8.1. sure or gravity, 25–30 L of fluid may be removed

Table 8.1 Complications of haemodialysis2,3,7,11

Complication Reason Treatment

Hypotension ‘crash’ Too much fluid being removed too quickly Assess dry weight (25–60%) e.g. if patients have come in significantly Perform sodium or ultrafiltration profiling over their dry weight Give at least 100 mL bolus of sodium Too many antihypertensives chloride 0.9% via the haemodialysis machine Dry weight too low and raise the patient’s feet above the head Autonomic neuropathy Reduce the rate of fluid removal Omit antihypertensives on dialysis days Stop the patient eating on dialysis Give midodrine 2.5–30 mg or fludrocortisone, carnitine 20 mg/kg (IV), sertraline 50–100 mg daily Cramps (5–25%) This can also be related to hypotension or if Give a bolus of 50–100 mL sodium chloride the patient has too much fluid removed 0.9% or quinine sulfate tablets (e.g. due to an incorrect dry weight) Reassess patient’s weight Reduced serum sodium usually near the end Give carnitine, vitamin E supplementation of dialysis Carnitine deficiency Pruritis (1–5%) Dry skin, hyperphosphataemia, uraemic Give antihistamines and moisturising lotions toxins or an allergic reaction to heparin or or change the dialyser the dialyser membrane Thrombosis Especially with dialysis catheters but also Usually give urokinase/alteplase locks or can occur with fistulas, usually if the infusions according to local policy. Urokinase haemoglobin is too high infusions can vary from 10 000 to 250 000 IU over 1–24 hours People with fistulas may be on prophylactic aspirin or low-dose warfarin Reduce/stop ESA and iron (continued overleaf) 92 Chapter 8 • Renal replacement therapy

Table 8.1 (continued)

Complication Reason Treatment

Infections Especially at the exit site of dialysis Low threshold for commencing patients on catheters or fistulas due to an increase in antibiotics, mainly anti-staphylococcal agents manipulations and skin penetration (e.g. flucloxacillin, vancomycin or Dialysis patients also have an impaired gentamicin). Continue for 2–3 weeks. immune response Antibiotic locks of gentamicin/heparin, The main organisms involved are Staph. vancomycin/heparin, taurolidine or citrate aureus or Staph. epidermidis. Rarely they may also be used can be due to Gram-negative organisms, May require replacement of catheter if it mainly with femoral haemodialysis becomes colonised catheters. They can be very serious and Nasal mupirocin may be used for nasal lead to infections like septic shock, Staph. carriers endocarditis or discitis To prevent hepatitis B, units should vaccinate Hepatitis B and C outbreaks can also very their patients and staff and any infected rarely occur patients should be isolated Isolate hepatitis C patients Screen for both viruses on a regular basis Anaemia Blood loss, inadequate dialysis, excessive Give ESA and intravenous iron bleeding post dialysis, iron or Reduce the heparin dose erythropoietin deficiency Hair loss Heparin Try low-molecular-weight heparin or one of Low zinc levels the other agents used for anticoagulation Zinc supplements Dialysis-related Minor effect of disequilibrium syndrome Give low-dose clonidine 25–75 µg pre- migraine/headache possibly due to excess urea removal dialysis (5–10%) Arrhythmias Can be linked with hypokalaemia Increase dialysate potassium concentration (5–60%) Nausea and Associated with hypotension and Give antiemetics vomiting (5–15%) disequilibrium syndrome Reduce blood flow rate at the start of dialysis. Recirculation This is the repeated uptake of blood which Access may need to be reviewed has already been dialysed. Usually leads to poor adequacy and/or biochemistry per treatment.2,18,19 Haemofiltration can be blood flow rate to help prevent this happening either continual or intermittent depending on or the blood flow rate can be increased. It does the stability of the patient, although continual is give a more accurate fluid balance as the best for very unstable patients. replacement fluid is returned after all the fluid The post-dilution method is most common has been removed at the filter. now but has problems with clotting if the blood Pre-dilution is slightly less efficient for becomes too concentrated in the filter due to a removal of larger molecular weight compounds reduction in blood flow. Therefore the ultra- than for small molecules; it is the best treatment filtration rate should be less than 20% of the if you have to remove more than 25 L a day or Continuous renal replacement therapies 93

Haemodialysis Flow scheme haemofiltration

Dialyser Anticoagulation Blood pump Sterile infusion solution

Ultrafiltrate Blood from the patient

An exact Blood to the patient balancing of infused and ultrafiltrated volumes is mandatory when using haemofiltration!

Figure 8.4 Haemofiltration circuit. Copyright and courtesy of Fresenius Medical Care.

␤ if the patient has a high haemoglobin as the 2-microglobulins, advanced glycation end- blood is diluted before the filter and therefore products). there is reduced clotting and the filter lasts • Continuous renal replacement therapy is longer. More fluid is needed compared with better for cardiovascular and intracranial post-dilution.18–20 stability, especially in patients at risk of cere- The advantages of convective and continu- bral oedema (e.g. after neurosurgery or acute ous treatment are as follows:2,3,21,22 liver failure) due to slower fluid shifts and a more gradual change in electrolytes. If fluid • It improves cardiovascular stability, possibly is removed too rapidly, as in intermittent due to convection producing a cooling effect haemodialysis, it would result in a reduction in the extracorporeal circuit and by the re- in cardiac filling pressures and cardiac insta- moval of inflammatory mediators and the bility. Body water is unable to move fast more biocompatible membranes that are used. enough from the intracellular and interstitial • It gives better control of blood pressure. spaces to the plasma.2,22 • More physiological fluid removal is possible due to a reduction in hypovolaemic effects. The disadvantages of convective and con- 11,22,23 • It can remove larger quantities of fluid (up to tinuous treatment are as follows: 3–6 L/day) as it is also usually carried out • It is more expensive than haemodialysis as over a longer time interval so there is a more fluid and more expensive filters are reduced risk of disequilibrium syndrome. required. Because of this it is used as a short- Also it can be used to create ‘space’ in fluid- term treatment in the acute situation. restricted patients who need enteral or • It is a more complex treatment so extensive parenteral nutrition or are receiving intra- training of staff is required. venous antibiotics. • There is no definite measure of adequacy as • It produces a higher rate of removal of yet. molecules with a middle (500–15 000 Da) • Small molecule clearance (e.g. urea) is or large (>15 000 Da) molecular weight (e.g. reduced. 94 Chapter 8 • Renal replacement therapy

Continuous arterio-venous haemofiltration can be removed gradually. Replacement fluid is not usually required. There can be problems Continuous arterio-venous haemofiltration though with lines clotting due to low blood (CAVHF) was the first haemodialysis system flow and haemoconcentration.2,11 developed in the 1960s. It uses the patient’s The dialysers used for haemofiltration are blood pressure to drive the filtration so no known as haemofilters and are high flux, usu- blood pumps are needed. Blood leaves by the ally synthetic hollow-fibre dialysers.2 They have arterial line and returns by the venous line. It to be very permeable to both fluids and solutes, had the disadvantage of a high risk of clotting as extensive ultrafiltration has to take place. in hypotensive patients and was not very There is significant clearance of both phosphate efficient – only between 10 and 20 L of plasma and amino acids; about 10% of intravenous water could be removed a day, so dialysis would amino acids from parenteral nutrition may be still sometimes be required to remove the small removed by haemofiltration.22 Biocompatibility molecules. It was good for anaemic patients and issues with the filters are similar to those for those with low protein levels as it depended on haemodialysis. the movement of plasma water i.e. the thinner Haemofiltration fluid is a balanced electro- the blood the easier the patient was to treat lyte solution of sodium, calcium, magnesium (similar to pre-dilution haemofiltration, when and chloride, which usually comes in 5 L bags. the replacement fluid dilutes the blood before The rate of haemofiltration fluid flow rate is the filter). usually 1–2 L/h increasing to 3–4 L/h for high- CAVHF then led to CAVHD (continuous volume haemofiltration with an ultrafiltration arterio-venous haemodialysis), which improved rate of at least 35 mL/kg/h.3 The rate is usually small solute clearance and resulted in better decided by how much fluid has to come off the cardiovascular stability than intermittent patient per day compared with how much fluid haemodialysis. CAVHDF (continuous arterio- to return to the patient. The fluid is available venous haemodiafiltration) is better still for with or without potassium, and 1–4 mmol/L of haemodynamically unstable patients and is potassium are usually added per bag if they are more efficient than CAVHD, but unless you potassium free; the amount added depends on have on-line fluid production it can be very the patient’s blood results. The fluid is usually expensive.11,19,22 buffered with lactate as lactate is metabolised to Pumped circuits were then developed which bicarbonate, which is used to correct the only needed venous access so removed some of patient’s underlying metabolic acidosis. If the problems associated with arterial access (e.g. patients have problems metabolising lactate elderly atherosclerotic patients who have poor (e.g. patients with hepatic failure, severe sepsis access). This is known as CVVHF (continuous or cardiogenic shock) then lactate-free or veno-venous haemofiltration). It is more effi- bicarbonate solutions should be used as they cient than CAVHF because the blood pumps can increase cardiovascular stability, improve controlled the ultrafiltration and so it can be dialysis symptoms (nausea, vomiting, headache, used in people with low blood pressure, but it is and hypotension) and patient well-being.2,22 not as good for haemodynamically unstable Anticoagulation has to be balanced with patients. CVVHD (continuous veno-venous excess bleeding in acute renal failure patients. If haemodialysis) improves efficiency once again, at all possible it is best to avoid anticoagulating and CVVHDF (continuous veno-venous the patient and just flush the lines with haemodiafiltration) improves efficiency and heparin, but this is not always possible. Heparin stability.11,19,22 is usually used, but if there are complications Slow continuous ultrafiltration (SCUF) usu- then epoprostenol can be used. It should be ally uses arterial access and can remove between noted that epoprostenol can cause severe 3 and 6 L of fluid a day. It is preferred for fluid- hypotension and the dose must be adjusted overloaded patients, such as patients with heart depending on the patient’s response; 40% of failure but without renal failure, as excess fluid epoprostenol will be removed by haemofiltra- Haemodiafiltration 95 tion. The other anticoagulants used in haemo- is known as a high-flux dialyser and has the dialysis may also be chosen, but heparin and ability to remove large volumes of extracellular epoprostenol are most commonly used.2,11,22 fluid and molecules up to 30 kDa. Replacement Access is mainly via dual or triple lumen cen- fluid and the filtered blood are then returned to tral lines inserted into the internal jugular or the patient.3,25 femoral vein for CVVHF/CVVHD/CVVHDF. The Dialysers that are used for haemodiafiltration lines are not usually tunnelled as they are norm- must have an ultrafiltration coefficient of at ally inserted at ward level so are generally least 55 mL/h/mmHg but this must be balanced changed every 4–5 days and can achieve a blood with excess albumin loss if the dialysers are too flow rate of around 150–300 mL/min.2 leaky. In order to make haemodiafiltration afford- able it is necessary to have on-line substitution fluid production from dialysis fluid. To do this Haemodiafiltration ultra pure water is required as the fluid goes directly to the patient. The water goes through Although the use of haemodialfiltration as a 2–3 extra filters compared with haemodialysis, long-term treatment is still in the early stages in one each for the water, the dialysate and the sub- the UK, it was first done in 1976 and is very stitution fluid and can be within the machine or common in Germany.24 Interest waned in the external depending on the machine.18 The high- 1980s, due partly to the cost of the replacement flux dialysers required can also be very expensive fluid compared with dialysis fluid and also but they are slowly coming down in price. because of developments in haemodialysis, such Advantages of haemodiafiltration are im- as bicarbonate dialysis, sodium profiling, accur- proved cardiovascular tolerance to fluid ate volume monitoring to allow weight loss to removal, removal of middle molecules, inflam- be controlled, all of which could improve matory markers and advanced glycation end- cardiovascular stability. Interest picked up again products (AGEs) and better blood pressure as people realised the long-term complications control.26,27 and increased mortality of haemodialysis due to One of the problems with haemodiafiltration the accumulation of middle and large-molecular- is the increased removal of drugs, especially ␤ weight molecules (e.g. 2-microglobulins) and vitamins, antibiotics and low-molecular-weight also with the ability to produce on-line heparins. This must be taken into consideration, haemofiltration fluid (also known as substitu- and if using low-molecular-weight heparins as ␤ tion or replacement fluid). 2-Microglobulin is anticoagulation for haemodiafiltration an addi- associated with complications such as amyl- tional dose is sometimes required half way oidosis and carpal tunnel syndrome. Some through the dialysis. Care must also be taken ␤ studies have shown a reduction of 70% in 2- with vancomycin, as up to 50% of vancomycin microglobulin, which may reduce these compli- may be lost during haemodiafiltration, necessit- cations of long-term dialysis.23,25–27 ating increased doses. Approximately 3.5% of patients on dialysis are on haemodiafiltration, and if high-flux dia- lysis (i.e.dialysis using high-flux dialysers but Efficiency no convection) is included the total is taken up to 25%. Efficiency can be affected by:24 As the name suggests, haemodiafiltration is a combination of haemodialysis and haemo- • Dialyser membrane and surface area filtration (simultaneous diffusion, ultrafiltration • Blood flow rate – QB (range is 300– 3 and convection). This gives clearance of small, 500 mL/min; an increase in QB can increase middle and large molecules. Blood is withdrawn small solute clearance) as for haemodialysis and passes through a very • Dialysis fluid flow rate – QD (range is 500– permeable dialyser where diffusion occurs. This 800 mL/min) 96 Chapter 8 • Renal replacement therapy

• Substitution fluid volume – QS (standard is • A good blood flow is required in order for it 60 mL/min, range 0–150 mL/min for post- to be efficient, or else the number of hours dilution and 0–250 mL/min for pre-dilution on dialysis must be increased ␤ and can affect 2-microglobulin removal) • More anticoagulation is required • Treatment duration • There may be problems with the lines clot- ␤ • Haemoglobin concentration ( 2-microglo- ting and efficiency if the haemoglobin is too bulin is mainly found in plasma and there is high and the blood is too viscous, as the an inverse correlation with its removal and blood concentrates in the dialyser the patient’s haemoglobin) • Treatment is of longer duration as small mol- • No one is quite sure how to measure ade- ecule clearance is not as good.24,28 quacy in haemodiafiltration as conventional There is some work being done at the measurements (e.g. Kt/V) only take small moment looking at mid-dilution haemodia- molecule clearance into account. filtration which gives better removal of the In pre-dilution haemodiafiltration the blood middle and large molecules but is worse for is diluted by the substitution fluid before going small molecules (e.g. urea and creatinine) through the dialyser. This was the original form compared with post-dilution. Here you get post- of haemodiafiltration. dilution occurring first and then pre-dilution Its advantages are that: within the dialyser. A special dialyser is required, which is split into two internally. • It is better tolerated than post-dilution Problems encountered with this method are haemodiafiltration increased albumin loss (equivalent to peritoneal • Less heparin is required dialysis), increased clotting and increased • Blood is less likely to clot in patients with a removal of low-molecular-weight heparins.28 good haemoglobin or access problems.21,26

The disadvantages of pre-dilution haemodi- afiltration are that: Peritoneal dialysis • Small solute clearance is reduced as the sub- stitution fluid has diluted the blood before Although the principle of peritoneal dialysis the diffusion has taken place in the dialyser had been considered as early as 1923, it was not so there is a reduced equilibrium gradient. until 1959 that it was first used because pro- Higher infusion rates of substitution fluid cedural problems held up development. In 1968 are possible to compensate for this (not too Tenckhoff and co-workers developed suitable important as small molecules will be access for peritoneal dialysis and then in 1976 removed by diffusion) Popovich and colleagues developed an ambu- • ␤ -Microglobulin clearance is reduced 2 latory dialysis system. It has had varying popu- • Approximately twice as much substitution larity due to consultant bias, haemodialysis fluid is needed compared with post-dilution availability and financial constraints.30 By the haemodiafiltration (i.e. higher dialysate flow end of 2002 there were 130 000 people on peri- rates are needed).18 toneal dialysis worldwide, representing 15% of Post-dilution haemodiafiltration is the more the total dialysis population.31 It is a good usual method now; blood is diluted by substi- choice for diabetic patients (as people with dia- tution fluid after going through the dialyser. betes tend to have more vascular access prob- The advantages with this method are that: lems and heparin on dialysis can increase the risk of retinopathy), children, people with • Smaller volumes of fluid are required unstable cardiovascular disease and those who • It is more efficient, getting the best removal are well motivated and still pass urine, or of small and large molecules. patients who live a significant distance from the The disadvantages are that: dialysis centre.30 Peritoneal dialysis 97

Peritoneal dialysis works on the three main and tunnelled through the abdominal wall to principles of diffusion, osmosis and convection. emerge on one side of the abdomen (15 cm of A solution is infused into the peritoneal cavity the catheter remains outside the body). The where the patient’s own peritoneum acts as the catheter can be inserted under general or local semi-permeable membrane. The peritoneal anaesthesia and can be put in laparoscopically. membrane is a thin, highly vascular, stretchable This has a success rate of 75% and the addition membrane, with a surface area of approximately of one or two Dacron cuffs helps to secure the 2m2. Diffusion and convection of solutes catheter in place by facilitating fibrous growth occurs between the capillary blood and the dia- in the tunnelled section, which helps reduce lysis solution in the peritoneal cavity, with infections.7,32 (Figure 8.5). excess fluid being removed by osmosis.30,32 It is advised that patients should receive There is less clearance of the small solutes prophylactic antistaphylococcal agents (e.g. (e.g. urea and creatinine) but this tends not to vancomycin or flucloxacillin) before insertion be too big a problem as the patients usually still of the catheter to reduce the risk of infection.33 pass urine.30 Some units may also advise a 5-day course of Peritoneal dialysis is good for initial therapy nasal mupirocin for people who are staphylo- as it preserves vascular access, there is a reduced coccal carriers. The catheters are usually not risk of contracting hepatitis B and C, reduced used for two weeks after insertion. During this cardiac dysfunction, reduced anaemia and doses time the exit site should be kept dry.33 It is also of erythropoiesis-stimulating agents (ESA); it is very important that the patient does not also less expensive than haemodialysis. become constipated either before the catheter is in situ or once it is in place, as that can then cause problems with the dialysis. Post care of Access the exit site is usually with a liquid antibacter- ial soap or Betadine or saline solutions.32 Access is via a small (6 mm in diameter), soft, Peritoneal dialysis is not as aggressive as flexible catheter (e.g. Tenckhoff catheter), usu- haemodialysis and does not leave the patient ally made of silicone rubber, inserted into the feeling washed out at the end of each session, peritoneum at the midline below the umbilicus as disequilibrium syndrome is rare compared

Extra-abdominal

Subcutaneous Preperitoneal cuff

Skin

Subcutaneous fat

Muscle and fascia

Preperitoneal fat

Peritoneum

Transmural Subcutaneous cuff

Intraperitoneal

Figure 8.5 Positioning of peritoneal dialysis catheter. Copyright and courtesy of Baxter Healthcare Ltd. 98 Chapter 8 • Renal replacement therapy with haemodialysis. It can also preserve residual advantage that it is easy to teach people how to renal function.30 Dietary and fluid restrictions use it as no machine is involved. tend not to be as strict as on haemodialysis The dialysate is warmed to body temperature because patients usually still pass some urine by bag warmers and run into the patient/ and because dialysis is done every day rather peritoneal cavity via the catheter by gravity (this than three times a week as with haemodialysis. usually takes about 10 minutes) and remains in Peritoneal dialysis is not recommended for the peritoneum for about 4 hours, known as the people who:32 dwell time. After 4 hours the patient connects their catheter to a drainage bag and a fresh bag • are homeless of fluid. The fluid in the peritoneal cavity is • have breathing problems drained out, this usually takes about 10–20 min- • have diverticular disease utes, and a fresh bag is then drained in. This is • have had previous abdominal surgery known as the ‘flush before fill’ system and can • have problems with body image reduce the risk of infections. The optimum • lack storage space for supplies dialysate fill volume that the body can tolerate • are anuric is 2.5 L for a 1.73 m2 body surface area; the aver- • have hernias. age volume used is 2 L (range 1.5–3 L). If the (These are not all necessarily contraindications.) body surface area is greater than 2 m2 then a fill The dialysate prescribed uses mainly glucose volume of 3–3.5 L can be tolerated. The volume as the osmotic agent and traditionally comes in of fill is reduced if there is a history of hernia, three strengths: weak, medium and strong. The pleural leak or if the patient experiences abdom- solution also contains lactate, sodium, potas- inal pain.30,32,34 sium, calcium and magnesium. Weaker solu- tions are used if the patient is dehydrated and Automated peritoneal dialysis stronger ones if the patient is fluid-overloaded. The consultant and peritoneal dialysis nurse This follows the same principles as CAPD except decide the prescription that is most suitable for the exchanges are done by a machine during the patient depending on the result of adequacy the night over 8–10 hours (usually six tests, fluid balance and the patient’s blood exchanges). In automated peritoneal dialysis results. The prescription may need to be (APD) 1.5–3 L of fluid are drained into the peri- changed during periods of peritonitis as the toneum by a machine, left for about 1–3 hours peritoneum becomes more permeable. when it is then drained out and fresh dialysate The characteristics of several commonly used drained in. This process is repeated throughout peritoneal dialysis fluids are summarised in the night until the total prescription volume Table 8.2. has been reached (e.g. 10 L). This is a more expensive procedure than CAPD but many patients find it more socially Types of peritoneal dialysis acceptable as the patient can lead a normal life during the day, free from dialysis exchanges. As There are two main forms of peritoneal dialysis: the dwell times are shorter than traditional CAPD, an extra bag of fluid may need to be run • Continuous ambulatory peritoneal dialysis in at the end of dialysis and kept in during the • Automated peritoneal dialysis. day to improve the efficiency of the dialysis. This can be either glucose or starch-based Continuous ambulatory peritoneal dialysis (icodextrin).34 Another advantage is that larger total fill vol- The patient usually performs this form of dia- umes can be used as the intraperitoneal pressure lysis manually 4–5 times each day although is reduced when lying down. This is good for carers can also be trained to do it. Continuous bigger patients as they can get more exchanges ambulatory peritoneal dialysis (CAPD) has the and for high transporters (see later) who tend to Peritoneal dialysis 99

Table 8.2 Characteristics of some peritoneal dialysis fluids31,35–37

Fluid Advantages Disadvantages

Basic glucose fluida (e.g. Good osmotic agent Weight gain Dianeal 1.36%, 2.27%, Can control ultrafiltration by changing Short-lived ultrafiltration (6 hours) 3.86%, Gambrosol 1.5%, the glucose concentration GDPs produced during heat sterilisation 2.5%, 3.9%) Cheap can increase the production of AGEs, which can damage the peritoneum and is associated with atherosclerotic cardiovascular disease in diabetics Increased insulin requirements Hyperlipidaemia Pain on filling due to low pH Glucose-based with Reduced pain on instillation Need to mix before using due to bicarbonate buffera (e.g. Reduced GDPs instability Physioneal, pH 7.4, More physiological Weight gain Balance, pH 7) Short-lived ultrafiltration (6 hours) Increased insulin requirements Amino acid-based (e.g. Maintains nutritional balance Not very efficient at improving nutrition Nutrineal, pH 6.7, 1.1% Can give 25% of daily protein intake Can only be used once a day due to amino acid solution, 15 Reduced weight gain from glucose increase in urea nitrogen concentrations amino acids (64% therefore better for diabetics and mild metabolic acidosis essential)) No GDPs No evidence it can prevent malnutrition Less hyperlipidaemia Starch-based polymers Longer ultrafiltration life (8–12 hours) Sterile peritonitis (e.g. icodextrin Improved diabetic control Rash (Extraneal)) Reduced weight gain from glucose Pain due to low pH (especially if patient therefore better for diabetics is used to the bicarbonate-based bags) Reduced GDPs Can only be used once a day due to Can be used to increase the adequacy maltose accumulation (unknown long-term of patients on APD if they require a problems) daytime exchange Both maltose and icodextrin metabolites Achieves better ultrafiltration than can give falsely high glucose levels in 2.27% glucose bag monitors that use dehydrogenase Reduced hyperlipidaemia pyrroloquinolinequinone Expensive

aDifferences in calcium concentration, usually 1.25 or 1.75mmol/L. Some bags are available as ‘specials’ with lower calcium and magnesium concentrations. GDP, glucose-degradation product; AGE, advanced glycation end-products. be fluid-overloaded with CAPD due to reduced about half of the fluid is drained out and ultrafiltration; this is overcome by the short replaced with clean fluid at each exchange, dwell times with APD. This form of treatment which occurs every 30–60 minutes. It can also can also reduce the incidence of peritonitis, as be used in patients who complain of pain with there are fewer connections to be made by the traditional APD. The high flow rate with this patient, so reducing the risk of contamination. form of treatment leads to better diffusion, as Tidal peritoneal dialysis is a variant of APD. the equilibrium gradient is always present.34 Dwell times are considerably shorter and only One of its disadvantages is that large volumes 100 Chapter 8 • Renal replacement therapy of dialysate are required, up to 30–36 L per tion rates over a longer period while reduc- session.32 ing the patient’s glucose exposure. Some factors that can affect adequacy are:

Adequacy • No residual renal function • Large body weight • Hypo- or hyper-permeable membranes Adequacy depends on the permeability and effi- • Overhydration ciency of the membrane, net ultrafiltration rate, • Peritonitis episodes. peritoneal blood flow rate and the dialysate flow rate and is related to small solute clearance. This The transport status of a patient (i.e. the per- is calculated as either a creatinine clearance or meability of their peritoneal membrane) is Kt/V estimation. Both of these take into account determined using a peritoneal equilibrium test any residual renal function as well as the clear- (PET), this can also give an indication as to ance from the dialysis. To measure adequacy whether the patient is best suited to APD or you need a 24-hour urine collection to measure CAPD. This will normally be measured at the urea and creatinine and also the volume of beginning of peritoneal dialysis (within 4–8 urine and a blood sample at the end of the col- weeks), if there have been a number of peri- lection. This gives an indication of the patient’s tonitis episodes that may have damaged the residual renal function. To calculate the clear- peritoneum, or if there have been a lot of ance from the dialysis you collect all the drain mechanical problems.5 samples for 24 hours and combine them, from this you take a 10 mL sample to measure urea, Procedure albumin, glucose and creatinine. You also need to measure the volume drained in and the vol- 1 Usually a 2.27%, 2 L bag is kept in overnight. ume drained out. Both of these measurements The next morning it is drained out and the are put into a computer program, which calcu- volume measured. lates the patient’s adequacy. It should be 2 Another bag of the same strength and vol- checked every six months, if the patient is fluid- ume is drained in with the patient lying overloaded or symptomatic and one month down, every 400 mL (usually drained in over after a regimen change. 2 minutes) the patient is rolled from side to The British Renal Association Standards side to ensure that the whole peritoneum is document states that renal units should aim for coated. a Kt/V greater than 1.7 and a creatinine clear- 3 Then 10 mL samples are taken at 0, 2 and 4 ance of at least 50 L/week/1.73 m2.5 In the USA, hours. This is done by withdrawing 200 mL the National Kidney Foundation KDOQI and taking 10 mL from that sample and Guidelines have suggested a target Kt/V of at returning the remaining 190 mL to the least 2 and a creatinine clearance greater than patient. These are sent to the lab to measure 60 L/week/1.73 m2.2,31 These targets are for for urea, creatinine and glucose. CAPD. Higher targets should be aimed for in the 4 A venous blood sample is taken at 2 hours to APD population. measure urea and electrolytes, bone profile Some methods of enhancing adequacy and glucose. are:30,31 5 After 4 hours, the dialysate is drained out and the volume measured. • Increase bag volumes for CAPD • Increase frequency of exchanges for CAPD This procedure measures the ratio between • Increase the strength of the bag dialysate to plasma (D/P) urea, creatinine and • Increase the dwell time glucose for a set period of time compared with • Use icodextrin during the day for APD or the volume drained out after 4 hours and is overnight for CAPD. This is a glucose poly- used to assess the loss of ultrafiltration, trans- mer product which produces high ultrafiltra- port status or under-dialysis. The D/P creatinine Peritoneal dialysis 101

catheter needing to be removed.5 There is a Table 8.3 Transport status supposedly reduced incidence with APD although this was not shown in an audit in High transporters Very efficient membrane Scotland between 1999 and 2002.35 Repeated Solutes are transported quickly infections can lead to damage to the peritoneal Increased glucose absorption membrane, leading to technique failure and Poor ultrafiltration hospital admission. Risk of hypoalbuminaemia Good hygiene is one of the most important Best on APD with shorter dwell factors to prevent exit site infections and peri- times tonitis. If the catheter is dropped or the pro- Low transporters Inefficient membrane Very good ultrafiltration tective cap falls off then prophylactic antibiotics Solutes are transported slowly should be given to prevent peritonitis, this is Best on CAPD with longer dwell usually a stat dose of vancomycin or a course of times oral flucloxacillin. The main causes of peritonitis are poor APD, automated peritoneal dialysis; CAPD, continuous ambulatory hygiene (usually due to a skin contaminant), peritoneal dialysis. exit site or tunnel infections, colonoscopy, dia- rrhoea, constipation or diverticular disease (due to movement of bowel bacteria – known as at 4 hours is used to classify the patient as a enteric peritonitis).33 ‘high’, ‘high average’, ‘low average’ or ‘low’ Peritonitis presents as abdominal pain, nau- transporter. Ten per cent of the population are sea, vomiting, diarrhoea, pyrexia, flu-like symp- high transporters, 53% high average, 31% low toms and cloudy peritoneal dialysis effluent average and 6% low (Table 8.3). bags, usually with a white cell count >100 mm3 Peritoneal dialysis tends to have a limited and at least 50% polymorphonuclear neutrophil survival, up to 8 years with only 20% remain- cells.7,33 ing at 10 years. Repeated peritonitis episodes White cell counts, culture sensitivities and account for 36% of dialysis failure, due to the Gram stains are sent off although antibiotics peritoneal membrane becoming fibrosed, adhe- should be started empirically while waiting for sions forming and it becoming inefficient for the results and changed if required. Antibiotics dialysis. These patients then transfer to the to cover both Gram-positive (vancomycin or a haemodialysis programme. ‘Burn out’ can be cephalosporin) and Gram-negative (aminogly- another problem in peritoneal dialysis as some cosides or third-generation cephalosporin, e.g. people find that they can only cope with it ceftazidime or oral ciprofloxacin) organisms for a few years before it becomes too much for should be started. If the bags are cloudy then them.31 500–1000 units/L of heparin should be added to each bag to dissolve the fibrin in the peri- toneum until the bags are clear. Complications of peritoneal dialysis It is possible to get culture-negative results sometimes due to the culture process or the The main complications with peritoneal dialysis patient may be on antibiotics. The Renal are associated with the insertion of the catheter Association states that you should have less and infections.33 than 15% of culture-negative episodes.5,33 The antibiotics are usually administered intraperitoneally (IP) together into the peri- Peritonitis toneal dialysis fluid bags, although penicillins The Renal Association standards state that less and aminoglycosides should not be mixed than one episode per 18 patient months is an together due to incompatibility. The antibiotics acceptable peritonitis rate for a unit with an should be added to a bag that will have a dwell initial cure rate greater than 80% without the time of at least 6 hours. 102 Chapter 8 • Renal replacement therapy

The antibiotics can be administered daily or site and peritonitis infections. This is advised with each exchange; vancomycin is usually only by the Renal Association.5,31,33 Methicillin- required weekly depending on levels. Very good resistant Staph. aureus (MRSA) is more diffi- systemic absorption is obtained from IP anti- cult to treat. biotics; some units monitor aminoglycoside levels but others do not. There are opposing These two Staph. species are the most com- views on whether the small amount of amino- mon organisms implicated in peritonitis; Gram- glycoside we give can affect the patient’s positive infections account for 75% of cases. residual renal function; some units use • Enterococcal infections (from the gastro- cephalosporins instead, although they may not intestinal tract) and streptococcal infections be as effective for peritonitis. The antibiotics are usually associated with a greater morbid- should be continued for at least 2–3 weeks ity and are often linked with exit site or depending on the severity of the infection. The tunnel infections. peritonitis will usually start to resolve after 48 • Gram-negative infections are usually caused hours. If the catheter has to come out it should by Escherichia coli, Klebsiella and Proteus be out for 6–8 weeks, but this may depend on organisms, and are usually transmural due to the patient and the medical and nursing staff. constipation, colitis, touch contamination or There are various terms linked with peritoni- exit site infections. Treatment is with 33 tis episodes: cephalosporins (e.g. ceftazidime); there is a • Refractory peritonitis: condition does not high incidence of treatment failure. improve after 5 days of antibiotics and usu- • Pseudomonas infections are usually due to ally will require the catheter to be removed skin contamination and are linked to exit site • Recurrent peritonitis: repeat episode within and tunnel infections. They are treated with four weeks with a different organism two different antibiotics as they can result in • Relapsing peritonitis: repeat episode within quite a severe peritonitis; one is usually a four weeks with the same organism or sterile quinolone and the other, for example, peritonitis ceftazidime. The catheter may need to be • Repeating peritonitis: repeat episode after removed. more than four weeks with the same • Mycobacterium tuberculosis or non-TB peritoni- organism. tis can be very difficult to diagnose. There is an increased incidence in Asia. It is treated Death is more likely with Gram-negative with conventional antituberculosis drugs.33 bacilli and fungi. The organisms associated with peritonitis include: Fungal peritonitis is usually preceded by pro- longed or multiple courses of antibiotics and • Coagulase-negative staphylococcus (CNS) has an incidence of 2–7%. Treatment in this (e.g. Staphylococcus epidermidis, which case is usually IP, IV or oral with either single or accounts for 50% of cases) is usually from combination treatment with amphotericin, touch contamination and is often quite fluconazole, flucytosine, voriconazole or caspo- straightforward to treat.30 It can even be fungin, depending on sensitivities and local pro- treated at home if the patient is taught how tocol. Usually the catheter needs to be removed. to administer the antibiotics to the peri- If the catheter is not removed quickly there is a toneal dialysis bags. high risk of mortality, with death occurring in • Staphylococcus aureus infections can be severe 25% of patients. Intraperitoneal amphotericin and are also due to touch contamination or can cause chemical peritonitis and is also quite more usually from exit site infections. painful on administration. However, when Infection is also associated with nasal car- amphotericin is given intravenously, there is riage of Staph. aureus and prophylactic nasal poor penetration into the peritoneum. With and/or topical mupirocin or cyclical flucytosine it is important to monitor levels to rifampicin have been shown to reduce exit limit the risk of bone marrow toxicity develop- Peritoneal dialysis 103 ing. Antifungal therapy should be continued for peritoneum by AGEs have been associated with 10 days after the catheter has been removed.33 sclerosing peritonitis. Sclerosing peritonitis is irreversible sclerosis Treatment can be with prednisolone at a dose of the peritoneal membrane and usually occurs of 0.5 mg/kg/day and tamoxifen 10–40 mg daily after patients have been on peritoneal dialysis and/or other anti-inflammatory or immuno- for a long period of time. It can be related to suppressive drugs (e.g. azathioprine) or surgery, continual inflammation, and the risk increases although there is no definite treatment.39,40 after 4 years on peritoneal dialysis. Sclerosing Removal of the catheter is essential and peri- peritonitis can be difficult to detect and CT and toneal dialysis must be discontinued, as it can ultrasound scans are required.38 Chlorhexidine be fatal. gluconate, which is sometimes used for cleaning Other complications of peritoneal dialysis are the exit site, and long-term damage to the listed in Table 8.4.

Table 8.4 Other complications of peritoneal dialysis2,7,11,29,31,33

Other Reason Treatment complications

Exit site infections Can be due to poor technique Treat with antibiotics for at least two weeks (usually Staph. Environmental (e.g. showers) (e.g. flucloxacillin for Staph. aureus or aureus or If not treated may result in tunnel infections, quinolones for Ps. aeruginosa) Pseudomonas peritonitis Hypertonic saline dressings may also be used infections) Diagnosis: purulent drainage from exit site, (1 tablespoon of salt to 500 mL water, apply erythema alone not necessarily an to gauze), wrap around catheter for 15 indication of infection minutes Enhanced training can reduce recurrent infections Prophylactic mupirocin or gentamicin cream may be used but there is a concern about resistance developing. (Mupirocin ointment should not be used as it may corrode polyurethane catheters) Staph nasal carriers may be given mupirocin nasal cream prophylactically Flow or drainage Constipation (can be due to drugs, reduced Laxatives. In severe cases drugs like Picolax problems with the motility or hypokalaemia which can reduce may be required catheter bowel motility) Replacement/manipulation of catheter in Catheter misplacement, migration or theatre kinking Urokinase lock of 5000 IU Fibrin deposition Add heparin 500 units/L to dialysis fluid for a few days Peritoneal leaks Catheter used too soon Rest the catheter and temporary Badly positioned catheter haemodialysis Can leak into tissues, lungs and scrotum In extreme cases catheter may require to be removed Technique/ultrafiltra- Due to repeated occurrences of peritonitis, Increase strength of bag but this can lead to tion failure (3% after this can cause damage to the peritoneum damage of the peritoneum 1 year, 30% after 6 leading to ultrafiltration failure and Reduce the dwell time years) inadequate dialysis Add in icodextrin Fibrosis due to AGEs Change to haemodialysis (continued overleaf) 104 Chapter 8 • Renal replacement therapy

Table 8.4 (continued)

Other Reason Treatment complications

Hernia Due to the force of the fluid in the abdomen Surgery and temporary haemodialysis Can cause flow problems if they are large Blood in dialysate If patients are menstruating Treat peritonitis Peritonitis present Investigate other causes Heparin is being added to the bags Strenuous exercise Malnutrition (severe Possibly associated with chronic Use an amino acid based dialysis fluid 8–10% and mild inflammation and is associated with poor Refer to a dietitian for supplements and 30%) prognosis, more research is needed in this advice area Due to albumin and amino acid loss in the dialysate, reduced intake and appetite, abdominal fullness, delayed gastric emptying and glucose absorption

AGE, advanced glycation end-products.

C CASE STUDIES

Q1. At the monthly multidisciplinary meeting, a haemodialysis patient’s urea reduction ratio is only 50%. What can be done to improve their adequacy?

Q2. Miss RB is an 80-year-old woman recently started on haemodialysis. After about an hour on dialysis she becomes hypotensive. What can be done about it?

Q3. Mr MP has recently started on CAPD and has been having problems with pain on draining in his fluid. What could be the reason for his pain and what dialysis fluid should he use?

Q4. Mr BA is a 45-year-old man on peritoneal dialysis. He was admitted with abdominal pain the previous evening. What are the possible causes of his abdominal pain and what should be done?

Q5. On checking the notes it appears that Mr BA has had a couple of peritonitis episodes in the past few months and is a known staph carrier. What would you recommend?

Q6. Miss RB did not have good enough veins for a fistula so has a permanent catheter in place. She has come in today and the line is not working. What can be done?

Q7. Mr MC has only been on haemodialysis a few weeks and it was noticed that his platelet count has fallen quite dramatically. Heparin-induced thrombocytopenia (HIT) has been diag- nosed. What can be used as an anticoagulant for his dialysis? References 105

References Sedgewick J, eds. Principles and Practice of Renal Nursing. London: Stanley Thornes, 1998: 74–89. 20. David S, Tagliavini D, Cambi V. Pre-post dilution 1. Fleming L. A view of dialysis through the ages. Br haemofiltration. Nephrol Dial Transplant 1989; 4: J Ren Med 1999/2000; 4: 20–22. 37–40. 2. Levy J, Morgan J, Brown E, eds. Oxford Handbook 21. David S, Cambi V. Haemofiltration: predilution of Dialysis. London: Oxford University Press, versus postdilution. Contrib Nephrol 1992; 96: 2004. 77–85. 3. Fraser D, Venning M. Principles of haemodialysis. 22. Kirby S, Davenport A. Haemofiltration/dialysis Medicine 1999; 27: 44–46. treatment in patients with acute renal failure. 4. Ringrose T. What’s new in vascular access? Br J Care Crit Ill 1996; 12: 54–58. Ren Med 1998; 3: 6–8. 23. Locatelli F, Di Filippo S, Manzoni C. Removal of 5. Renal Association and Royal College of small and middle molecules by convective tech- Physicians of London. Treatment of Adult Patients niques. Nephrol Dial Transplant 2000; 15 (Suppl and Children with Renal Failure: Standards and 2): 37–44. Audit Measures, 3rd edn. Renal Association 24. Wizemann V, Kulz M, Techert F et al. Efficacy of and Royal College of Physicians of London, haemodiafiltration. Nephrol Dial Transplant 2001; 2002. 16 (Suppl 4): 27–30. 6. Rodger RSC, Briggs JD. Renal replacement ther- 25. Ledebo I. Principles and practice of hemofiltra- apy in the elderly. Scot Med J 1997; 42: 143–144. tion and hemodiafiltration. Artif Organs 1998; 22: 7. Stein A, Wild J, Cook P, eds. Vital Nephrology. 20–25. London: Class Health, 2004. 26. Canaud B, Bosc JY, Leray-Moragues A et al. On- 8. Chesser AMS, Baker LRI. Temporary venous line haemodiafiltration. Safety and efficacy in access for first dialysis is common, undesirable long-term clinical practice. Nephrol Dial Trans- and usually avoidable. Kidney Int 1997; 52: 267. 9. Hoenich N. Choosing a haemodialyser. Br J Ren plant 2000; 15 (Suppl 1): 60–67. Med 1997; 2: 15–18. 27. Padrini R, Canova C, Conz P et al. Convective 10. Hoenich N, Graham K. Membranes for renal and adsorptive removal of beta2-microglobulin replacement therapy. Br J Ren Med 1996; 1: during predilutional and postdilutional haemo- 18–21. filtration. Kidney Int 2005; 68: 2331–2337. 11. Daugirdas JT, Blake PG, Ing TS, eds. Handbook of 28. Krieter DH, Falkenhain S, Chalabi L et al. Clinical Dialysis, 3rd edn. Philadelphia: Lippincott cross-over comparison of mid-dilution hemodia- Williams & Wilkins, 2001. filtration using a novel dialyzer concept and post- 12. Shaldon S. Dialyser reuse: A practice that should dilution hemodiafiltration. Kidney Int 2005; 67: be abandoned. Semin Dialysis 1993; 6: 11–12. 349–356. 13. Lowrie EG, Zhensheng L, Ofsthun N et al. 29. Gokal R. Peritoneal dialysis and complications of Reprocessing dialysers for multiple uses: recent technique. In: Davison M, Cameron AM, analysis of death risks for patients. Nephrol Dial Grunfield et al., eds. Oxford Textbook of Clinical Transplant 2004; 19: 2823–2830. Nephrology, 2nd edn. London: Oxford University 14. Tielemanns C, Madhoun P, Lenaers M et al. Press, 1998. Anaphylactoid reactions during hemodialysis on 30. Gokal R. Peritoneal dialysis. Medicine 1999; 27: AN 69 membranes in patients receiving ACE 47–49. inhibitors. Kidney Int 1990; 38: 982–984. 31. Gokal R. Peritoneal dialysis in the 21st century: 15. Hoenich N. Quality of water in dialysis. Br J Ren An analysis of current problems and future devel- Med 1999; 4: 21–22. opments. J Am Soc Nephrol 2002; 13: S104–S116 16. Ashley C, Currie A. The Renal Drug Handbook, 2nd 32. Graham C. Principles of peritoneal dialysis. In: edn. Oxford: Radcliffe Medical Press, 2004. Challinor P, Sedgewick J, eds. Principles and 17. Peticlerc T, Jacobs C. Dialysis sodium concen- Practice of Renal Nursing. London: Stanley tration: what is optimal and can it be indi- Thornes, 1998: 167–183. vidualized. Nephron Dial Transplant 1995; 97: 33. Piraino B, Bailie GR, Bernardini J et al. ISPD 596–599. Guidelines/Recommendations: Peritoneal dialysis- 18. Ledebo I. On-line hemodiafiltration: Technique related infections recommendations: 2005 and therapy. Adv Ren Replace Ther 1999; 6: update. Perit Dial Int 2005; 25: 107–131. 195–208. 34. Williams P. Automated peritoneal dialysis – new 19. Challinor P. Acute renal failure. In: Challinor P, targets. Br J Ren Med 1997; 2: 17–19. 106 Chapter 8 • Renal replacement therapy

35. Kavanagh D, Prescott GJ, Mactier RA. Peritoneal peritonitis: identification of diagnostic, clinical, dialysis-associated peritonitis in Scotland (1999– and radiological features. Am J Kidney Dis 1994; 2002). Nephrol Dial Transplant 2004; 19: 24: 819–825. 2584–2591. 39. Del Peso G, Bajo MA, Aguilera A et al. Clinical 36. Marshall J, Jennings P, Scott A et al. Glycaemic experience with tamoxifen in peritoneal fibrosing control in diabetic CAPD patients assessed by syndromes. Adv Perit Dial 2003; 19: 32–35. continuous glucose monitoring system (CGMS). 40. Evrenkaya TR, Atasoyu EM, Unver S et al. Kidney Int 2003; 64: 1480–1486. Corticosteroid and tamoxifen therapy in scleros- 37. Dasgupto MK. Strategies for managing diabetic ing encapsulating peritonitis in a patient on patients on peritoneal dialysis. Adv Perit Dial continuous peritoneal dialysis. Nephrol Dial 2004; 20: 200–202. Transplant 2004; 19: 2423–2424. 38. Campbell S, Clarke P, Hawley C et al. Sclerosing 9

Renal transplantation

Andrea Devaney and Mark Lee

Replacing a failed organ of the body with globulins. However, despite the marked another from a human or animal donor has improvement seen in one-year graft survival long been desired as an effective method of with current immunosuppressive regimens, the curing disease. The first report of a human renal incidence of graft loss over time remains sig- transplant was in 1936.1 However, in the nificant.4 This is principally due to chronic absence of effective immunosuppressive agents, allograft nephropathy (CAN). It is also the case organs were rapidly rejected unless the donor that most recipients die with a functioning was an identical twin to the recipient.2 graft, typically from cardiovascular causes. Thus, Allografts (hereafter termed grafts) are organs the focus over the last decade has shifted from from non-identical members of the same reducing the frequency of acute rejection to species. Several series of allograft transplants minimising both CAN and patients’ cardio- were reported in the 1950s with the advent of vascular risks. methods of suppressing the immune system. Renal transplantation is significantly more Only after the introduction of azathioprine in cost effective to the health service than any 1961 did one-year graft survival rates signifi- form of dialysis and offers a patient the oppor- cantly improve. Azathioprine was usually used tunity of a normal lifestyle.5,6 There are not in combination with corticosteroid. Anti- enough donor kidneys, however, to fulfil lymphocyte globulins also became available in demand: 5773 patients were registered on the the 1960s and were used both in short courses UK transplant waiting list for a renal transplant around the time of transplant (induction as of April 2006. This has increased by around immunosuppression) and to treat the frequent one-third between 1996 and 2005 and now rejection episodes. Co-morbidities were signifi- represents almost 100 patients per million of cant from the high doses of induction and population.7 maintenance immunosuppression. It was not until the introduction of ciclosporin in the early 1980s that both graft The donor organ and patient one-year survival rates dramatically improved to figures comparable with today’s clinical practice (one-year graft survival of Transplant organs are obtained either from a 85–95% and patient survival of 95–100%). cadaveric donor (heart beating or non-heart Maintenance triple therapy with ciclosporin beating/asystolic) or a living donor (related or combined with lower doses of corticosteroid unrelated). Recipients receive a solitary kidney and azathioprine became the standard until the unless, rarely, the donor is a small child and introduction of tacrolimus, mycophenolate and there is not a suitable paediatric recipient. In sirolimus in the 1990s.3 this case both donor organs may be trans- The late 1990s saw the launch of engineered planted into the adult recipient in an en bloc monoclonal antibodies as alternative induction operation. Some diabetic patients with end agents to the non-specific anti-thymocyte stage renal disease (ESRD) will receive a

107 108 Chapter 9 • Renal transplantation simultaneous kidney and pancreas transplant negative, then the other probable cause of early from a cadaveric donor which will also cure non-function is usually surgical, for example their diabetes. A patient’s native kidneys are left renal artery thrombosis or a tamponade result- in situ unless they have previously been ing from a ureteric or blood vessel anastomotic removed (for example because of recurrent leak. infection, hypertension or, in the case of cystic kidneys, to make space for the transplant). The graft is usually placed extraperitoneally in Transplant rejection and its prevention the right ileac fossa for ease of surgical access to the iliac arteries, veins and the bladder. Most patients are transplanted whilst stabil- Transplant rejection is mainly the result of ised on dialysis; however, some patients may responses by the recipient’s adaptive immune receive a pre-emptive transplant, usually within system once donor antigens on the kidney have 6–12 months of their anticipated need for dia- been recognised as non-self. Typically the lysis. With the fall in numbers of cadaver patient will feel unwell with a temperature and organs, living donor transplant programmes tenderness over the kidney, urine output will have expanded in the UK and this now repre- diminish and serum creatinine will rise by more sents one-third of the total kidney transplant than 25% over 24–48 hours. The effect is medi- activity. Kidney transplants currently account ated by the recipient’s T- and B-lymphocytes. for four out of every five solid transplants in the Some of the potential for rejection can be min- UK, but despite efforts to increase donation, imised by carefully matching donor and recipi- overall activity remains static (Table 9.1). 7 ent characteristics (human leukocyte antigens (HLAs) and ABO blood group antigens are the most relevant) but immunosuppressive medi- Graft function cines, taken vigilantly for the life of the graft, are the mainstay of prevention. Urine volumes and daily measurement of a Ensuring ABO blood group compatibility is a recipient’s serum creatinine levels are the stan- matter of routine across all transplanted organ dard ways of monitoring transplanted graft types to prevent hyperacute rejection. function. The incidence of delayed graft func- Hyperacute rejection, which ensues within min- tion (DGF), where the recipient continues to utes and hours of unclamping the blood vessels require dialysis for a period after the operation, to the graft, will irreversibly damage the kidney, is higher with cadaveric grafts, particularly leading to its loss if a recipient has pre-formed those from asystolic donors. The term DGF is antibodies to donor antigens. In addition to usually reserved for the temporary, recoverable ABO blood group incompatibility a recipient situation when the kidney presents histologic- might have pre-formed antibodies from expos- ally with acute tubular necrosis in the absence ure to non-self antigens (for example HLAs) of signs or symptoms of other causes of early from a previous blood transfusion, transplant or non-function. Assuming that the cross-match is pregnancy. To prevent this a cross-match of

Table 9.1 Comparative kidney transplant numbers for the UK 2003–2006

Transplant 2003–2004 2004–2005 2005–2006

Total kidneys 1849 (65%) 1783 (65%) 1799 (82%) Total pancreas (including 59 (2%) 86 (3%) 126 (6%) kidney and pancreas) Total solid organ transplants 2867 2724 2196 Transplant immunosuppression 109 donor lymphocytes with recipient serum is the same for the donor and recipient. That there undertaken when the recipient is called for are, for example, over 120 HLA-DR antigens surgery and a negative result is required for the alone makes clear the need for the national transplant to proceed. registration and donor kidney allocation Acute rejection is most commonly seen in scheme as undertaken by the UK Transplant the first few months after a transplant but can Special NHS Health Authority. In practice, few occur at any time. The standard is for diagnosis cadaveric grafts are HLA identical and the total to be confirmed by histological examination number of mismatches is taken into account of core biopsy tissue, which often shows T- when deciding the immunosuppressive regimen lymphocyte infiltration underlining the import- for that patient. ance of this type of response in rejection. The Chronic allograft nephropathy is also some- Banff classification is the histological standard times termed chronic rejection. It can present as for excluding or the differential diagnosis of the early as just a few months post transplant, but type and severity of rejection.8 is most common after a number of years. To understand how acute rejection can be Compared with acute rejection, the progress of prevented both by HLA matching and the deterioration of kidney function is slow and action of immunosuppressive drugs, a mech- largely relentless. The aetiology is not under- anistic overview of the provoked immune stood but there are thought to be many factors response is needed. that contribute to the response (for example Donor antigens on the surface of graft cells cytomegalovirus infection and the nephrotoxic- (for example, HLA-A) are presented to resting T- ity of calcineurin inhibitors). Episodes of acute helper (Th) lymphocytes by macrophages and rejection are risk factors for the subsequent other antigen-presenting cells (APCs). The development of chronic rejection.9,10 recognition process activates the Th-cells to pro- duce and secrete chemical messenger cytokines (e.g. interleukin 2 (IL-2)) and activate cell sur- Transplant immunosuppression face receptors for them (e.g. IL-2R). This line of T-cells now differentiates and proliferates under the influence of the chemical messengers. To maintain longevity of the transplant, Cytotoxic Tc-cells are activated and begin to lyse patients are required to take immunosuppress- the donor cells. At the same time B-lymphocytes ive drugs to attenuate the natural immune are activated by cytokines, eventually leading to response to the allograft. Immunosuppressive antibody formation against the antigen. The drugs can be categorised by pharmacological intense, localised immune activity prompts class or by when and how they are used. the involvement of the innate immune system Induction therapy refers to those drugs used with its phagocytes and enzyme systems, such peri-operatively (for example high-dose, intra- as ‘complement’. In higher grade acute vascular venous corticosteroid or a short course of rejections a greater degree of antibody deposi- IL-2R-blocking antibodies). Maintenance im- tion is seen. Untreated acute rejection results in munosuppression usually consists of two or graft loss within a few days. three drugs with different and complimentary Reduced rates of rejection and improved mechanisms of action at lower individual long-term graft survival is seen if the ‘foreign- dosages to achieve a cumulatively adequate ness’ of donor kidney cells to the recipient is immunosuppression. It is usual to aim for reduced by histocompatibility testing and greater exposure to maintenance immuno- matching. Humans inherit two antigens from suppression in the first few months after a trans- each of the HLA-A, -B and -DR groups, one plant (for example, higher corticosteroid doses haplotype from each parent. An HLA identical or higher target tacrolimus trough levels). The match, sometimes called a zero mismatch, will secondary aim of the maintenance regimen is to only be present if all six specific HLA antigens minimise individual drug adverse effects, (two HLA-A, two HLA-B and two HLA-DR) are including their contribution to cardiovascular 110 Chapter 9 • Renal transplantation risk, and the risk of infection and malignancy Thymoglobulin), the humanised anti-inter- which strongly correlates with the degree of leukin-2 receptor (IL-2R) antibodies basiliximab overall immunosuppression. and daclizumab, alemtuzumab and rituximab. The third category of immunosuppression is Each antibody will be discussed in turn. that used to treat acute rejection should it occur. The first-line treatment is usually a short course Rabbit anti-thymocyte globulin (rATG, of high-dose, intravenous corticosteroid. Acute Thymoglobulin) rejection can also be resolved by changing the maintenance immunosuppression and this Thymoglobulin remains unlicensed in the UK. might be undertaken alongside pulsed steroid. It is a rabbit polyclonal antibody directed at a Histologically higher grade rejections might wide variety of human T-cell surface antigens, require a course of antithymocyte globulin (for including major histocompatibility complex example, Thymoglobulin) or muromonab CD3 (MHC) antigens and adhesion molecules. It is a (OKT3), either first line or if a pulse of cortico- depleting antibody and thus confers long- steroid fails. lasting immunosuppression. During the first year post transplant, graft Some units use it as induction on a daily survival rates are similar for most modern basis for a short course of specified duration. immunosuppressive regimens. All transplant Other centres choose to administer doses on an centres will have their own preferred induction intermittent basis with the decision to dose and maintenance regimens for different scen- being based on CD3+ count, total lymphocyte arios. Unless agreed otherwise with local com- count or absolute T-cell count. One study found missioners, regimens in the UK will be National patients receiving daily ATG had a longer and Institute of Health and Clinical Excellence more profound duration of lymphocyte deple- (NICE) compliant.12 There is a tension between tion than those receiving intermittent dosing.11 applying a consistent, standardised and audit- A test dose of 5 mg infused over 1 hour is given able approach and yet still tailoring the choice prior to administration of the first full dose to of drugs to the collective assessment of donor check for any sensitivity to rabbit protein. If a and recipient risk factors. Some of the many fac- patient suffers an untoward reaction during the tors that might affect the choice of drug include test dose then this contraindicates any further the donor’s age, organ origin, the recipient’s treatment with Thymoglobulin. The usual daily age, ethnicity, concordance and transplant his- dose is 1.25–2.5 mg/kg. Central venous admin- tory, titre of antibodies against a standard panel istration, by a slow infusion over 6–12 hours, is (panel reactive antibodies or PRA) and the preferred as phlebitis and inflammation can be degree of HLA mismatching. a problem; however, there is growing experience Long-term patient and graft survival is the with peripheral administration. Intravenous new Holy Grail of transplantation. The goal of corticosteroids, antihistamine and oral paracet- maintenance immunosuppression is to optimise amol should be given 30 minutes before admin- these outcomes and to help maintain a high istration of the first full dose, and subsequent quality of life. doses if necessary, to reduce the cytokine release syndrome which most patients experience to a greater or lesser degree. Induction immunosuppression Basiliximab, daclizumab Much research continues around induction ther- apy in an effort to find the ideal regimen to These antibodies were approved by the NICE minimise or avoid maintenance steroids and cal- review on immunosuppression12 as an option cineurin inhibitors with the goal of improving for induction therapy as part of a calcineurin long-term outcomes in renal transplantation. inhibitor-based immunosuppressive regimen. Several induction antibody agents are available, Both are monoclonal human/mouse antibodies including: rabbit anti-thymocyte globulin (rATG, manipulated to disguise the mouse region and Transplant immunosuppression 111 so minimise antibody formation in the recipi- be utilised with this agent remain to be ent. They bind very specifically to a part of the determined. IL-2R found only on the surface of activated T- lymphocytes and reduce the sensitivity of the Rituximab receptor to the cytokine IL-2. Unlike ATG they do not affect the inactivated or resting T-cell Rituximab is unlicensed in transplantation. It is population. The effect persists for 2–3 months. a high-affinity mouse–human chimeric mono- NICE specified that the drug with the lowest clonal antibody directed against the human acquisition cost should be used and most units anti-CD20 antigen, which is a cell surface recep- have elected to use basiliximab on this basis. A tor found mostly on B-lymphocytes. It inhibits phase IV trial evaluated the effectiveness and B-cell proliferation while inducing cellular safety of basiliximab when added to a regimen apoptosis. It may reduce pre-existing PRA titres consisting of ciclosporin (Neoral), azathioprine and has been shown to assist in overcoming and corticosteroids.13 When compared with positive cross-matches in planned ‘incompat- placebo, and as observed in other trials,14 the ible’ transplants. It has a long-lasting duration basiliximab group showed a lower incidence of of action. A recent report17 found that 15 acute rejection without increasing adverse months after a single induction dose of ritux- effects, notably infection or post-transplant imab, B-cells were still considerably reduced in lymphoproliferative disease (PTLD). One-year peripheral blood and there was complete elim- patient and graft survival rates were similar in ination within the graft. The exact place of the two groups. These findings were corrob- rituximab in immunosuppressive therapy is yet orated in a 2004 meta-analysis involving 38 to be established, however, and again there is a trials that enrolled nearly 5000 patients.15 need for controlled trials to support its use in No trials to date have directly compared transplantation. basiliximab and daclizumab. Individually they have similar patient and graft outcomes but they have different pharmacokinetic profiles Maintenance immunosuppression and, at licensed doses, differ in cost and fre- The optimal maintenance immunosuppressive quency of administration. therapy in renal transplantation has not been established. Conventional maintenance regi- Alemtuzumab mens consist of a combination of agents that differ by mechanism of action. This strategy Alemtuzumab is unlicensed in transplantation. minimises the morbidity associated with each It is a humanised monoclonal antibody directed drug while maximising effectiveness. By the against the CD52 cell surface receptor that is early 1990s a combination of ciclosporin, expressed on most T- and B-lymphocytes, azathioprine and steroid was accepted as the eosinophils and on some populations of mono- standard maintenance regimen. We now have cytes, macrophages and dendritic cells. Like more drugs and many more possible combina- ATG it is a depleting antibody and confers tions and so preferred regimens vary between long-lasting immunosuppression. It is used in each UK transplant centre and indeed across the induction with the aim of allowing steroid-free world. The major immunosuppressive agents and/or calcineurin-free/sparing maintenance that are currently used in various combinations immunosuppressive protocols. A recent system- are: atic review of alemtuzumab’s use in organ trans- plantation concluded there is a need for more • Corticosteroids (primarily oral prednisolone) and larger randomised trials, with long-term • Antimetabolites, such as azathioprine, follow-up, before its role can be established in mycophenolate mofetil (CellCept), myco- practice.16 The dose, route and frequency of phenolate sodium (Myfortic) administration of alemtuzumab, and optimal • Calcineurin inhibitors, such as ciclosporin, maintenance immunosuppressive regimen to tacrolimus 112 Chapter 9 • Renal transplantation

• mTOR inhibitors, such as sirolimus, steroids have more known specific effects such everolimus as inhibiting interleukin 1, preventing T- • Investigational maintenance immunosup- lymphocyte proliferation and altering lympho- pression agents. cyte response to antigen at an intracellular level. Initial regimens will involve doses of predni- The NICE appraisal on immunosuppressive solone 15–20 mg/day which will then be therapy in renal transplantation in adults12 reduced to a lower maintenance dose (e.g. made specific recommendations about when 5 mg/day) over a few weeks and months. In certain agents should be used. They acknowl- some cases recipients may be weaned off steroid edged that some of these recommendations where no acute rejection episodes have would result in medicines being prescribed out- occurred, rejection risks are lower and steroid side the terms of their marketing licence: co-morbidity is an issue (for example, hyperten- • Tacrolimus could be used as an alternative to sion or new onset diabetes). The long-term side- ciclosporin based on the relative importance effects of corticosteroids are diverse and well of each drug’s side-effect profile for indi- known. Patients maintained on even low-dose vidual people steroids for longer than 6–12 months should • Mycophenolate should only be used have their bone mineral density assessed and will often require treatment with bisphosphon- – where there is proven intolerance to cal- ates and adjunctive calcium and vitamin D to cineurin inhibitors, such as nephrotoxic- reduce the risk of fractures from the associated ity leading to risk of chronic allograft osteoporosis. dysfunction, and – where there is a very high risk of nephro- toxicity necessitating minimisation or Antimetabolites – azathioprine, avoidance of a calcineurin inhibitor mycophenolate mofetil (CellCept), mycophenolate sodium (Myfortic) • Sirolimus should only be used in cases of proven intolerance to calcineurin inhibitors Azathioprine is a precursor of 6-mercaptopurine, (including nephrotoxicity) necessitating which is further metabolised to thioguanine complete withdrawal of these treatments. nucleotides which disrupt cellular DNA and RNA production, prevent mitosis and so in- For the purposes of this chapter we will pro- hibit the proliferation of activated T- and vide information on each immunosuppressive B-lymphocytes. agent rather than the variety and efficacy of the Mycophenolic acid is the active moiety of numerous drug combinations. Suffice to say in both the mofetil salt and the enteric-coated the UK, unless medicines are being used in the sodium salt. It also impairs lymphocyte pro- context of a clinical trial, the combinations used liferation by blocking purine biosynthesis will generally be compliant with the NICE through reversible inhibition of the enzyme guidelines. Most transplant centres still use a inosine monophosphate dehydrogenase. triple therapy immunosuppressive regimen with Given current evidence, azathioprine and a calcineurin inhibitor, an antimetabolite and mycophenolate mofetil when used with micro- prednisolone. An element of individualisation emulsion ciclosporin appear to be similar in may occur, taking into account specific patient terms of acute rejection and medium-term graft characteristics. Many Trusts audit against their survival rates.18 Analysis of the large US Renal guidelines. Transplant Registry database indicates that patients prescribed mycophenolate mofetil experience a significantly lower incidence of Corticosteroids – prednisolone CAN compared with those taking azathio- The effects of corticosteroids in transplantation prine.19 Further prospective studies are required are complex and multiple. In addition to power- to determine whether outcomes such as long- ful, general anti-inflammatory properties, term allograft survival differ with these anti- Transplant immunosuppression 113 metabolites. Current UK practice is that mediated T-cell receptor signal transduction and mycophenolates are used de novo primarily in inhibition of IL-2 transcription. By inhibiting patients at higher risk of acute rejection (re- cytokine gene transcription, they suppress T-cell transplants, high PRA or immunological cause and T-cell-dependent B-cell activation. They of renal disease). Mycophenolate presentations differ in the target cytoplasmic protein are markedly more expensive than azathioprine (immunophilin) they bind to: ciclosporin binds and it is safe to assume that economic issues will to ciclophilin and tacrolimus to FKBP-12. play a larger role in choosing between the two. Tacrolimus is the preferred calcineurin inhibitor, The usual starting dose of azathioprine is principally because several studies have reported 1–2 mg/kg once daily. Mycophenolate mofetil is that it is associated with fewer acute rejection started at 1 g twice daily if the patient is on con- episodes than ciclosporin and is possibly associ- comitant ciclosporin. If, however, the baseline ated with improved allograft survival.20,21 In immunosuppression is tacrolimus or sirolimus, addition, it does not cause the cosmetic adverse the starting dose is often reduced to 500– effects associated with ciclosporin (e.g. hirsut- 750 mg twice daily as ciclosporin is known to ism and gum hypertrophy), which are known inhibit the entero-hepatic recirculation of the to effect compliance. Tacrolimus is more likely active metabolite of mycophenolate. In terms of to increase the risk of post-transplant diabetes active mycophenolic acid 1 g of mycophenolate mellitus, which may then result in decreased mofetil is the near molar equivalent to 720 mg allograft survival among affected patients. of mycophenolate sodium. Both agents are nephrotoxic and this is one Leucopenia is one of the most common side- known contributing factor to CAN. Doses are effects for both drugs and may necessitate a dose initiated on a weight basis and then adjusted reduction. Anaemia may occur more frequently according to blood trough levels as shown in with mycophenolate than with azathioprine. Table 9.2. Because of the pharmacokinetics of Excluding infection, the most frequent adverse tacrolimus (t = 12–14 hours), it is prudent to effects associated with the mycophenolates are only measure blood levels a maximum of three gastrointestinal, usually manifested as nausea, times a week when a patient is in hospital. gastritis and diarrhoea. There is not yet con- Tacrolimus is more water soluble than vincing evidence of any significant difference in ciclosporin and is not dependent upon bile salts gastrointestinal side-effects between the mofetil for absorption. However food intake can reduce salt and the enteric-coated sodium salt. Myco- the extent and rate of absorption of tacrolimus. phenolic acid blood levels can be measured as a In practice, patients are advised to either con- means of establishing the optimum dose of sistently take tacrolimus with food or away from mycophenolate, however the evidence for the food. Most patients choose to take it with food, usefulness of this is inconclusive. at the same time as all their other medications. On days when patients come to hospital for a blood test they are asked to not take their morn- Calcineurin inhibitors ing dose of calcineurin inhibitor. Instead they Both ciclosporin and tacrolimus are calcineurin should take all their other medication at the inhibitors. As such, their immunosuppressive usual time but bring their morning dose of cal- action is mediated via blockade of calcineurin- cineurin inhibitor to hospital with them and

Table 9.2 Calcineurin inhibitor initial doses and target blood levels

Calcineurin inhibitor Common initial oral doses Typical target blood levels (ng/mL) doses (mg/kg twice daily) 0–6 months Over 6 months

Ciclosporin (Neoral) 4 150–300 75–150 Tacrolimus 0.05–0.1 10–15 5–10 114 Chapter 9 • Renal transplantation take it after their blood test. This is to ensure the works on mTOR to inhibit the activation of pro- blood test taken is a trough level. tein kinase S6 (p70S6k), interfering with protein Reported difference in toxicity with synthesis and preventing cell cycle progression tacrolimus as compared to ciclosporin include: from the G1 phase to the S phase. The main advantage of this class of drugs is • More prominent neurological side-effects, that they are not nephrotoxic. In practice, such as tremor and headache sirolimus is used mostly outside of its licensed • More frequent incidence of post-transplant indication.22 This is largely due to its adverse diabetes mellitus effects profile, which is dissimilar to that of • Less frequent incidence of hirsutism, gingival other agents. hyperplasia and hypertension Early use of sirolimus, as described in the • More frequent diarrhoea, dyspepsia and licensed indication, can impair wound-healing, vomiting with dehiscence and breakdown of anastamoses • More frequent alopecia described. Given the high incidence of lympho- • Increased predisposition to polyoma virus cele formation there is a strong case for delay- infection. ing initiation of sirolimus until after the early post-operative period. Acne type rashes, mouth Generic ciclosporin ulceration and peripheral oedema are particu- Although not yet available in the UK at the time larly common and may be severe enough to of writing, generic ciclosporin will probably be warrant drug withdrawal. Hyperlipidaemia licensed in the near future. Ciclosporin is a crit- appears to be dose related and most patients ical dose drug and absorption can be highly taking sirolimus will require treatment with a dependent upon formulation in some patients. statin. Arthralgia and interstitial lung disease It has a narrow therapeutic index and the will generally be an indication for this drug’s potential consequences of a variance in bio- withdrawal. Anaemia and thrombocytopenia availability associated with unplanned generic are more commonly seen than leucopenia or substitution are very significant (e.g. graft rejec- neutropenia and particularly if sirolimus is com- tion or nephrotoxicity). To this end the British bined with another antiproliferative drug such National Formulary (BNF) states that ‘because of as mycophenolate. differences in bioavailability, the brand of The majority of sirolimus side-effects are ciclosporin to be dispensed should be specified related to greater exposure to the drug. Many by the prescriber’. units now aim for maintenance trough levels of 5–8 ng/mL, which are about one-quarter of the initial target ranges in phase III trials. Inhibitors of mammalian target of rapamycin NICE recommends that sirolimus is only (mTOR) used for proven cases of calcineurin inhibitor Sirolimus and everolimus are the two agents in intolerance, where either ciclosporin or this, the newest class of immunosuppressants. tacrolimus have to be withdrawn.12 The poss- At the time of writing, only sirolimus is cur- ible niche for sirolimus is in patients where rently available in the UK. These two drugs have the calcineurin inhibitor component of the a unique mechanism of action and add another immunosuppressive regimen is beginning to therapeutic category to the immunosuppression compromise renal function.23 armamentarium. Sirolimus and everolimus do Sirolimus has a long half-life (about 60 not share a mechanism of action with cal- hours24). There is controversy as to whether or cineurin inhibitors but, like tacrolimus, they are not a loading dose is necessary when therapy only active after binding to FKBP-12. Rather is initiated. Supporters believe a loading dose is than inhibiting IL-2 production they work required to ensure that the patient reaches further downstream to block cytokine-driven steady state quickly. Opponents believe the proliferation of T-cells, B-cells and vascular high peak levels associated with loading dose smooth muscle cells. The drug–FKBP complex can worsen early side-effects and tolerability. Transplant immunosuppression 115

Sirolimus and ciclosporin taken together inter- Acute rejection act to increase exposure to, and raise serum Methylprednisolone is used first line in the levels of, sirolimus. The drugs should be given treatment of acute rejection. It is given as an IV at least 4 hours apart until ciclosporin is with- infusion on a daily ‘pulse’ basis for 3 days, usu- drawn.25 There is no similar interaction with ally at a dose between 500 mg and 1 g/day. Its tacrolimus and sirolimus. Monitoring sirolimus anti-inflammatory action profoundly alters the trough levels is not required as frequently as effector phases of graft rejection, including with calcineurin inhibitors as steady state will macrophage function. As a means of com- take 10–14 days. Doses often need to increase parison, 500 mg methylprednisolone is an once the calcineurin inhibitor is eliminated equivalent anti-inflammatory dose to 625 mg and are adjusted according to target serum prednisolone. Patients often experience dyspep- levels. In common with the calcineurin sia and become emotionally labile over the inhibitors, sirolimus is metabolised by cyto- period of the pulse. Diabetic patients receiving chrome P450 3A4 and is subject to many drug a course of methylprednisolone will require an interactions which are detailed later in the IV insulin sliding scale in order to normalise chapter. their blood sugars. At the end of the methyl- prednisolone course, oral prednisolone may be Experimental maintenance immunosuppression added or the baseline maintenance dose increased for a short time as a further additional The only noteworthy drug being pursued in increase in overall immunosuppressive load. clinical trials at present is belatacept. This is a Maintenance immunosuppression may also selective blocker of CD28 co-stimulation. Co- be altered (e.g. replacing ciclosporin with stimulation is required for full activation of T- tacrolimus or azathioprine with mycophenol- cells by the antigen on the antigen presenting ate) usually alongside a course of high-dose cell. Belatacept marks a new approach to intravenous steroids or antibody therapy. maintenance immunosuppression as it is only Steroid-resistant acute rejection is so named available as an intravenous formulation. In because it refers to on-going rejection despite current phase II clinical trials maintenance one or more courses of high-dose, IV steroids. doses are administered every 4 or 8 weeks as a Anti-T-cell antibody therapy, such as rabbit anti- 30-minute intravenous infusion. Preliminary thymocyte globulin (already described) or 12-month data from an on-going study report muromonab-CD3 (OKT3), may then be used. that belatacept did not appear to be inferior to There is controversy about how to manage ciclosporin as a means of preventing acute rejec- maintenance immunosuppression during con- tion after renal transplantation.26 All patients comitant antibody therapy. Some advocate stop- received induction therapy with basiliximab, ping all maintenance immunosuppression and mycophenolate mofetil and corticosteroids. then re-introducing calcineurin inhibitor a few Patients were randomised to receive one of two days before the end of the antibody course. doses (low/high intensity) of belatacept or Others would support continuing maintenance ciclosporin (control arm). Belatacept may pre- immunosuppression unchanged throughout. serve the glomerular filtration rate (GFR) and Muromonab-CD3 (OKT3) is unlicensed in reduce the rate of chronic allograft nephropathy the UK. It is a mouse-derived monoclonal anti- and, if licensed, the challenge will be to incor- body directed against the CD3 antigen complex porate regular intravenous infusions in the renal found on all mature human T-cells. No test dose units transplant follow-up. is necessary. However, patients should be pre- medicated with 500 mg IV methylprednisolone Treatment of rejection prior to the first dose to reduce the cytokine release syndrome. It is given as a daily peri- pheral bolus dose over a 7–14 day period Treatment of rejection is influenced by the depending on patient’s response. Some units histological findings on the biopsy. 116 Chapter 9 • Renal transplantation administer muromonab-CD3 intermittently when switching to sirolimus to ensure that the according to absolute lymphocyte count. GFR is greater than 40 mL/min. Patients with a Antibody-mediated rejection (AMR or GFR less than 40 mL/min experienced a higher humoral or vascular rejection) is estimated to rate of serious adverse events in clinical trials. occur in 3–10% of all renal transplants.27 It is now a more clearly defined phenomenon and its detection is classically based on three key Drug interactions factors: characteristic histological findings (severe vascular changes), positive C4d staining Most of the immunosuppressants can be on the biopsy and the presence of donor- adversely affected by other drugs and some can, specific antibodies (DSA). There are emerging themselves, interact with common medicines. case reports describing numerous treatment The importance of interactions with immuno- options for AMR but none are clinically proven. suppression results from these drugs having a The options include: plasma exchange, intra- relatively narrow therapeutic window where venous immunoglobulin, immunoabsorption, excess exposure leads rapidly to adverse effect antilymphocyte therapy (anti-T-cell: Thymo- and reduced exposure risks acute rejection. globulin, anti-B-cell: rituximab), altered Use of live vaccines (e.g. measles, mumps and maintenance immunosuppression (usually to rubella (MMR), yellow fever or bacille Calmette- combination of tacrolimus and mycophenolate) Guérin (BCG)) is contraindicated in immuno- and finally a combination of those above. suppressed individuals. This is particularly important when considering paediatric recipi- ents and those planning more exotic foreign Chronic allograft nephropathy travel. It is also possible that patients will Chronic allograft nephropathy (CAN) was exhibit diminished response to killed and poly- referred to previously as chronic rejection or saccharide vaccines. chronic renal allograft dysfunction or transplant Colestyramine, magnesium and aluminium glomerulopathy. Unfortunately there are no are all known to reduce absorption of mycophe- universally accepted diagnostic criteria for CAN nolate. Agents that interfere with enterohepatic but there are characteristic histological changes recirculation should, where possible, be present on biopsy. The clinical diagnosis is avoided. As described earlier, replacing ciclo- usually suggested by gradual deterioration of sporin with tacrolimus is believed to increase renal function over time, increasing proteinuria enterohepatic recirculation of mycophenolic and worsening hypertension. The causes of CAN acid, increasing exposure to the active drug by are multifactorial and include both immuno- about 30%. Taking some immunosuppressive logic (e.g. cell-mediated immune responses, drugs with food is known to alter the rate and incidence of acute rejection, presence of growth extent of absorption, although the general factors such as transforming growth factor-␤) advice for renal transplant patients is to be con- and non-immunologic factors (e.g. occurrence sistent about dose timing with respect to meals of delayed graft function, on-going hyperten- rather than complicating otherwise difficult regi- sion, hyperlipidaemia, cytomegalovirus (CMV) mens. A potassium-rich diet should be avoided infection). and potassium-conserving medicines should be The prevention and management of CAN used cautiously in patients taking calcineurin remains one of the major challenges. inhibitors, in particular tacrolimus which has a Intervention usually results in changes to propensity to lead to hyperkalaemia. immunosuppressive medication. Usual practice The major categories of immunosuppressant would be stop the calcineurin inhibitor and interaction are those that disrupt metabolism. maintain patients either on a mycophenolate or Xanthine oxidase activity is inhibited by allo- sirolimus based regimen. There is no clinical purinol, slowing conversion of biologically evidence to support one drug being better than active 6-thioinosinic acid to its inactive meta- the other. However caution should be exercised bolite. Thioinosinic acid is a metabolite of Post-transplant complications 117

Care should also be taken when combining Table 9.3 The effect of metabolic interactions on other nephrotoxic drugs such as amino- calcineurin inhibitor and m-TOR inhibitor levels glycosides, NSAIDs and amphotericin with calcineurin inhibitors. Additive neurological Interacting drug Effect on effects are more likely to be seen when drugs blood level with such adverse effects are combined with Erythromycin and clarithromycin Increased tacrolimus. Myelosuppression is also more Diltiazem, nicardipine, verapamil Increased likely when drugs known to depress bone Fluconazole, itraconazole, Increased marrow function are used with the anti- ketoconazole, voriconazole metabolites. Rifampicin Decreased Lipid-lowering agents tend to cause problems Carbamazepine Decreased in practice. Many patients with kidney trans- Phenobarbital Decreased plants require concomitant use of HMG-CoA Phenytoin Decreased reductase inhibitors (statins) either to treat St John’s wort Decreased hypercholesterolaemia or as prevention of cardiovascular morbidity. It must be borne in mind that ciclosporin can increase the risk of azathioprine, so when allopurinol is given con- muscle toxicity of statins, simvastatin in par- comitantly with azathioprine, the dose of the ticular. A maximum daily dose of 10 mg sim- latter should be reduced to one-quarter of vastatin is recommended in combination. the original. Ciclosporin, tacrolimus and Ciclosporin also greatly increases exposure to sirolimus are metabolised by the cytochrome ezetimibe, though the significance of this is as P450 3A4 family of isoenzymes. If known in- yet unclear. ducers or inhibitors (Table 9.3) cannot be Interactions are not confined to licensed avoided then the immunosuppressant level medicines. Grapefruit juice will inhibit the must be frequently monitored and the dose metabolism of tacrolimus, ciclosporin and adjusted to maintain a safe effect. sirolimus, leading to greater exposure. The Tacrolimus is itself known to inhibit P450 herbal remedy St John’s wort is a potent enzyme 3A4 and care should be taken when other inducer and can rapidly reduce plasma levels of medicines metabolised by the liver are co- the aforementioned drugs. Echinacea is claimed prescribed (for example, oral contraception). to stimulate the immune system, which would Ciclosporin has been demonstrated to increase be counterintuitive for a patient on immuno- the exposure to concomitant diclofenac, a non- suppression. General advice for transplant steroidal anti-inflammatory drug (NSAID) with recipients is to avoid additional herbal remedies a high degree of first-pass metabolism. It has unless the safety is well established. been recommended that the dose of diclofenac is halved in combination with ciclosporin. In practice all NSAIDs (including topical) are usu- Post-transplant complications ally avoided in renal transplant recipients as they can impair function. A risk/benefit assess- ment must be made before they are used in any The patient’s state of health before the trans- circumstance and renal function monitored plant, and especially that of their cardiovascular closely if they are deemed necessary. system, is the key to a long-term successful Probenecid and other drugs which might outcome. However the consequences of surgery compete for excretion with the glucuronide and immunosuppression per se are far from metabolite of mycophenolic acid (for example, benign and offer many potential complica- aciclovir) should be used cautiously where a tions. The management of these in terms of patient has impaired renal function and where prevention, prophylaxis and treatment is dis- tubular secretion is likely to account for a sig- cussed below. Adverse effects of the indi- nificant proportion of the clearance. vidual immunosuppressants, whilst clearly a 118 Chapter 9 • Renal transplantation complication, are discussed earlier in the chap- Cytomegalovirus ter and not revised below. This virus from the Herpes family is common in Surgical complications tend to occur early, the general population, and in the developed often during the transplant admission. They can world four out of five have been exposed to it include such seemingly contradictory problems by the age of 60. It usually causes minor flu-like as disastrous renal artery thrombosis and life- illness, but infection in immunosuppressed threatening bleeding from the graft site. Most patients can cause life-threatening disease. patients will receive prophylaxis for graft Those most at risk of disease are recipients naïve thrombosis with either lower doses of low- to the virus (CMV negative) whose primary molecular-weight heparins (e.g. enoxaparin infection is from the organ of a CMV-positive 20 mg daily) or low-dose aspirin (e.g 75 mg donor. It is UK practice that such recipients daily). Lymphoceles and collections requiring either receive prophylactic treatment against drainage are not uncommon and recipients CMV or are monitored, pre-emptively for early occasionally develop a temporary, unilateral leg signs of developing infection and then treated. oedema caused by impaired lymphatic drainage CMV-positive recipients risk reactivation of secondary to compression by the transplanted their own virus or super-infection with a differ- kidney. Urethral catheterisation for the first few ent strain from the donor and prophylaxis or days after surgery relieves pressure on the pre-emptive treatment is often offered if the ureter/bladder anastamosis. Ureteric strictures immunosuppressive load is high (for example, if can largely be avoided by the placement of a rATG has been used). The agents commonly double J stent for the first few weeks. used for prophylaxis are aciclovir, valaciclovir and valganciclovir, the latter two being licensed Infection for this indication. Prophylaxis is usually given for around the first three months post trans- plant. All three drugs are renally excreted and Some immunosuppressants are associated with dose adjustment according to changing renal higher rates of infection than others but such function is crucial28. complications should be considered as a func- CMV disease can be delayed and occur at the tion of the overall immunosuppressive load the end of the prophylaxis. Signs and symptoms of patient has received since the transplant. This disease include rigors, night-sweats, pneumon- will increase the risk of bacterial, viral, fungal itis and deranged renal and liver function. and parasitic disease. Disease usually responds to antiviral therapy Bacterial urinary tract infection (UTI) is com- with intravenous ganciclovir. A clinical trial is mon, particularly whilst a ureteric stent is in on-going into the use of oral valganciclovir to situ, and most patients receive some antibiotic treat CMV disease. prophylaxis in the early months after surgery. UTI in an immunocompromised patient often requires longer than 3 days of antibiotic treat- BK virus ment. Trimethoprim can be used empirically but resistance is not uncommon where a patient BK virus is so-named after the initials of the is already taking co-trimoxazole prophylaxis. It patient from whom it was first isolated. It is a should also be noted that use of trimethoprim member of the polyomavirus family and, can itself elevate serum creatinine. although prevalent in humans, it appears to Life-threatening fungal infections such as only cause disease in immunocompromised aspergillosis are rare in renal transplant recipi- individuals. Primary infection usually occurs in ents. Much more common are Candida infections childhood (via oral and/or respiratory exposure) of the mouth, oesophagus and genito-urinary and once infected the virus remains dormant in tract. Many units will offer prophylaxis in the renal epithelium and lymphoid cells. Clinical early period post transplant, when the immuno- manifestations in renal transplant recipients suppressive burden is greatest. include: asymptomatic haematuria, haemor- Post-transplant complications 119 rhagic and non-haemorrhagic cystitis, ureteric in renal transplant recipients than in non- stenosis and tubulo-interstitial nephritis. Some immunosuppressed individuals.29 Patients with units now perform BK virus screening of urine a history of TB or thought to be at risk of reac- and blood. BK viral DNA is first detected in the tivation will generally be prescribed a course of urine followed by the plasma and finally the isoniazid 200–300 mg daily for between 6 and kidney. BK virus infection in transplant recipi- 12 months after the surgery. In such patients, ents is directly related to the patient’s immuno- who have often received a suboptimal diet suppressive load rather than use of any one before transplant, it is necessary to co-prescribe specific agent. Optimum management is pyridoxine 10 mg daily to prevent peripheral unproven but the usual first-line measure is to neuropathies. reduce immunosuppression. The goal of de- creased immunosuppression is to minimise viral replication without triggering rejection. There Malignancy are reports of alternative approaches in the literature, including treatment with intravenous Long-term immunosuppression confers on immunoglobulins, cidofovir or leflunomide. transplant recipients a greater risk of certain malignancies than the general population. A recent analysis of over 35 000 American renal Pneumocystis carinii pneumonia transplant recipients provides us with the Pneumocystis jiroveci (Pneumocystis carinii) is a largest experience on the relative incidence and parasitic organism which invades and infects types of malignancy in this population.30 It the alveolar space in the immunosuppressed. should be considered when reviewing this data Most of the Western population is exposed to P. that American immunosuppressive protocols are jiroveci by the age of 10 but only the immuno- typically more aggressive than European proto- compromised develop life-threatening pneumo- cols. The analysis showed: nia. Many transplant units in the UK prescribe • A twofold increase in common tumours co-trimoxazole 480–960 mg daily for at least the including colon, lung, prostate, stomach, period whilst induction and early maintenance pancreas, oesophagus, ovary and breast doses are providing a high immunosuppressive • A threefold increase in testicular and bladder burden. Those intolerant of co-trimoxazole can cancer receive intermittent nebulised pentamidine ise- • A fivefold increase in melanoma, leukaemia, tionate or daily dapsone. hepato-bilary, cervical and vulvo-vaginal In active infection there may be as many as cancers 109 organisms per gram of lung tissue, prevent- • A fifteenfold increase in renal cell carcinoma ing oxygen transfer and leading to hypoxaemia. • A twentyfold increase in non-melanoma skin Signs are mainly tachypnoea and hypoxia, with cancers, Kaposi’s sarcoma and non-Hodgkin’s approximately half of patients developing a lymphoma. fever and/or cough. Prompt treatment with high-dose, intravenous co-trimoxazole, aug- The British Association of Dermatologists has mented with high-dose oral corticosteroid, is designed a helpful patient information leaflet required if severe disease is not to be fatal. specifically for recipients with an organ trans- plant.31 This leaflet addresses the three key issues of skin cancer: early detection, early treat- Tuberculosis ment and ways to decrease the risk. Reactivation of latent tuberculosis (TB) is a par- ticular risk for those recipients from commun- Post-transplant lymphoproliferative disorders ities where this infection is endemic (e.g. black African and South-East Asian communities in Post-transplant lymphoproliferative disorders Britain). The relative risk of developing active (PTLDs) are often the most serious and poten- TB has been judged to be 37 times higher tially fatal complication of long-term immuno- 120 Chapter 9 • Renal transplantation suppression. In 2003, Andreone et al.32 reported and is a widely disseminated herpesvirus. The the incidence of PTLD to be 1%, with an over- human host cells for EBV are limited to B- all increasing frequency, which is 30–50 times lymphocytes, T-lymphocytes, epithelial cells higher than in the general population. The and myocytes. The majority of PTLDs are EBV- main contributing factors for development are driven B-cell lymphomas. Once diagnosed, the immunosuppressive load and the Epstein– there are a number of possible treatment Barr virus (EBV) status of the donor/recipient options. The usual first step is to reduce the (seropositive donor/seronegative recipient con- immunosuppression in a step-wise manner, ferring greatest risk). EBV is the primary agent monitoring the patient for signs of tumour of infectious mononucleosis (glandular fever) regression (shrinkage of lesions, reduced EBV

Table 9.4 Common changes to prescribed medicines on transplant

Pre-transplant medication Action

Antihypertensives (e.g. diuretics, ACE inhibitors Stop immediately pre-transplant and only re-introduce if BP and angiotensin II receptor blockers, calcium persistently >130/80 mmHg channel blockers, alpha-blockers) Beta-blockers and other anti-anginals (e.g. Continue (rebound tachycardia and angina risk). Review nitrates and nicorandil) indication and dose post transplant Statins, fibrates and ezetimibe Review indication, dose and possible drug interactions. Where appropriate continue Antiplatelets and oral anticoagulants Temporarily withhold and re-introduce before discharge. Some patients will require full anticoagulation with unfractionated heparin (easily reversed if biopsy necessary) Erythropoietins and iron Stop pre-transplant Phosphate binders Stop pre-transplant Vitamin D metabolites Continue if previous parathyroidectomy – review serum calcium daily (dose increase ± calcium supplement often necessary) Stop for all other indications Quinine preparations Stop pre-transplant Hypnotics and agents for restless leg syndrome Continue with a view to withdrawing slowly after discharge (e.g. clonazepam, gabapentin) Enzyme-inducing antiepileptics (e.g. Continue. Empirically double starting dose of calcineurin carbamazepine, phenytoin) inhibitor and monitor levels closely Other antiepileptics and drugs for neuropathic Continue and consider need for dose increases where drug is pain (e.g. gabapentin, pregabalin) renally cleared Antidepressants Continue Allopurinol Stop pre-transplant unless cause of renal failure familial hyperuricaemia (rare). If continued with azathioprine—reduce azathioprine dose by half and monitor WCC closely. Consider using mycophenolate instead

Gastro-protection (e.g. H2 antagonists, protein Clarify indication and continue post-transplant pump inhibitors) Insulin and oral hypoglycaemics Early post-transplant sliding scale insulin and re-instate with regular review of blood sugars (dose may need to increase) The pharmacist‘s role for transplant patients 121 viral load). If there is poor response or addi- immunosuppression the need for adjunctive tional therapy is required (based on the severity medication (for example, early use of calcium of the disease) then chemotherapy may be and vitamin D ± bisphosphonates) should be administered.33 Anti-B cell antibody therapy considered, but care should be taken to ensure with rituximab may also be indicated.34 that it is suitable for long-term concordance. Promoting adherence to the medicine regi- men is one of the most important roles for the renal pharmacist. We know that the prevalence The pharmacist’s role in caring for of 20–32% non-adherence in renal transplant transplant patients recipients is directly related to the incidence of late acute rejection and graft loss.35 Pharmacists Most of the surgical transplant units have access can work to reduce this by informing recipients to a specialist pharmacist who will take respons- about their medicines to the level they desire ibility for medicines management issues peri- (verbally and with written information) and operatively. Likewise the renal pharmacists at responding positively to self-reported side- secondary referral hospitals will see significant effects and concerns. Where possible, limiting numbers of transplant recipients, usually as out- medicine-taking to twice daily, tagging this to patients. Pharmacists’ remits will differ from another life function such as eating and provid- centre to centre and the roles mentioned below ing a medicine record prompt card can be facil- should be considered as a guide only. itated by the pharmacist working within the At the time of transplant all medications larger team. should be critically reviewed. Analgesia (e.g. Like concordance issues, avoidance of drug morphine PCA), regular aperients and throm- interactions is an ongoing role and many bosis prophylaxis will be prescribed and are cru- patients will contact their hospital pharmacist cial for an acceptable and safe inpatient before taking any prescribed or over-the-counter experience. Many medicines that are essential medicines. At the very least all recipients should for good health whilst approaching or receiving have a point of contact for advice. This is likely dialysis will no longer be required and may to extend beyond patients to other healthcare be discontinued. Some agents will need to be professionals with many pharmacists actively weaned slowly to prevent withdrawal effects. managing either ongoing immunosuppression There will be an enduring need for others, prescribing/supply or shared-care across the sometimes at higher dosage, sometimes tem- primary/secondary care interface. porarily withheld. Table 9.4 offers some guid- Transplantation is the most cost-effective ance on typical medicine changes made at the treatment option for chronic kidney disease time of transplant. (CKD). The CKD patient pathway aims to Vigilance is required during the early post- optimise the patient’s general health at all time transplant period of improving renal function points and this is continued with a transplant. and stabilisation of immunosuppressant dosing With modern immunosuppression, early acute to ensure medicine use is safe, effective and rejection rates are low, one-year patient and evidence-based. If there is DGF, and continued graft survival rates are excellent but the biggest dialysis is required, then the pharmacist must cause of graft loss is death with a functioning consider the timing of the newly prescribed graft. This, and management of chronic allo- drugs around this and, occasionally, their fluid graft nephropathy constitute the current Holy volumes. The need for some antimicrobial pro- Grail in transplantation. Over coming years phylaxis (e.g. CMV disease prevention) may not eagerly awaited long-term (>5 years) data on be immediately clear and may require some immunosuppressive therapies will be published. research (e.g. with the UKT Core Donor Details There also exists a dichotomy between too form held by the transplant coordinators and little immunsuppression predisposing to rejec- the patient’s transplant CMV IgG status). tion and too much immunosuppression causing With the newly prescribed regimen of infection and malignancy. This situation is 122 Chapter 9 • Renal transplantation carefully balanced for each individual patient, use of medicines and should be an integral taking into account donor and recipient details member of a multidisciplinary team involved in and the merits/negatives of each immuno- the care of transplant patients. In addition, the suppressive agent. pharmacist has a positive role in educating The renal pharmacist can make a vital con- the patient to aid long-term concordance and tribution on safe, effective and evidence-based adherence.

C CASE STUDY

JL is a 60 kg, 45-year-old, white, British woman diagnosed with insulin-dependent diabetes mellitus (DM) at the age of 17. She is treated for hypertension and is registered as blind secondary to diabetic retinopathy. Her IDDM has recently been very well controlled (HbA1c 6.4% last month). She is single and has three children, the youngest of whom was delivered by caesarean section 10 years previously, and after which she received a blood transfusion. She started peritoneal dial- ysis 4 years ago but, following two episodes of peritonitis which required peritoneal dialysis catheter removal, she is now on haemodialysis. She passes less than 50 mL of urine from her native kidneys each day. She was CMV negative when listed with UK Transplant 38 months ago.

Q1. What recipient factors may have led to JL remaining on the waiting list for over 3 years? JL is called for a kidney transplant. At this point she takes the following medicines each day: • Calcium acetate 1 g three times a day with meals (phosphate binder) • Alfacalcidol 0.5 µg daily (secondary hyperparathyroidism) • Vitamin B Co Forte 2 daily (supplements) • Folic acid 5 mg daily • Aspirin dispersible 75 mg daily (primary cardiovascular prevention) • Simvastatin 40 mg at night • Ramipril 5 mg daily (hypertension) • Epoetin beta 1000 units SC twice weekly (anaemia) • Carbamazepine 200 mg twice daily (neuropathic pain) • Insulin glargine 28 units SC each evening (IDDM) • Insulin lispro 6–10 units SC with meals.

Q2. Which medicines should be stopped at this point and which would normally continue to be prescribed? On day 0 she receives a CMV-positive renal transplant from a 29-year-old, cadaver donor to which she has a 2,1,1 HLA mismatch. She receives methylprednisolone 1 g in theatre. The trans- planted kidney perfuses well on the operating table but does not have primary function. Urine output is minimal (<4 mL/h). Ultrasound scans show the graft is perfused and there are no obvious surgical complications. JL is prescribed an immunosuppression regimen of tacrolimus, azathioprine and prednisolone. Her prescribed medicines are as follows: • Aspirin dispersible 75 mg once daily (currently withheld) • Simvastatin 40 mg at night → The pharmacist‘s role for transplant patients 123

C CASE STUDY (continued)

• Carbamazepine 200 mg twice daily • Enoxaparin 20 mg SC once daily • Morphine sulfate 50 mg/50 mL IV 1 mg bolus patient-controlled analagesia with 5-minute lockout • Paracetamol 1 g four times daily • Senna 15 mg at night • Human soluble insulin 50 units/50 mL IV sliding scale based upon hourly blood sugar • Tacrolimus 3 mg twice daily • Azathioprine 100 mg once daily • Prednisolone 20 mg in the morning • Co-trimoxazole 480 mg at night • Cefradine 500 mg IV twice daily for 2 days post-op.

Q3. What other antirejection strategy might have been considered? She requires haemodialysis on day 1, continues to be effectively anuric and on day 3 her tacrolimus trough level is 6 ng/mL.

Q4. Is this tacrolimus level usually considered adequate?

Q5. What might be contributing to this level? Her tacrolimus dose is doubled to 6 mg twice daily. She has further dialysis on day 3 and day 5. On day 6 her tacrolimus level is 12 ng/mL. She is now back on regular SC insulin albeit requiring higher doses. On day 6 she passes more urine (80 mL/24hr) and on day 7 more again (400 mL/24 h). For the first time her plasma creatinine level has not risen significantly over the previous day. Medicines discharge training is commenced.

Q6. What input can the pharmacist make to maximise the chances of medicine regimen adher- ence? On day 8 her urine output slows, she spikes a temperature of 38.1ºC and is tender over her left iliac fossa. JL’s transplanted kidney is biopsied under ultrasound guidance and, before the result is available, she is given an infusion of methylprednisolone 500 mg. A ‘sliding scale’ IV insulin infusion is prescribed simultaneously and the regular SC insulin prescription is stopped. Biopsy results the following day suggest a low-grade acute cellular rejection and a further 2 days’ doses of methylprednisolone 500 mg infusion are prescribed. Day 9 tacrolimus trough levels are 18 ng/mL and the dose is reduced to 4.5 mg twice daily.

Q7. If the transplanted kidney continues to function poorly what could be the possible reasons?

Q8. What treatment options would be typical? On day 11 JL’s urine output has increased to 1.5 L and her creatinine level has fallen from 450 to 375 µmol/L.

(continued overleaf) 124 Chapter 9 • Renal transplantation

C CASE STUDY (continued)

By day 13 her creatinine has fallen to 198 µmol/L and she is passing 2 L urine each day, her tacrolimus level is 12 ng/mL, blood sugars are controlled on SC basal/bolus insulin and she is deemed medically ready for discharge with thrice weekly follow-up.

Q9. In addition to co-trimoxazole, what other prophylaxis might you expect to see on the TTA?

References 12. National Institute for Health and Clinical Excellence (NICE). Technology appraisal 85: Immunosuppressive therapy for renal transplan- 1. Voronoy YY. El siglo Med 1936; 97: 296. tation in adults; September 2004. http://www. 2. Merrill JP, Murray JE, Harrison JH. Successful nice.org.uk/TA085guidance (accessed 15 July homotransplantations of the human kidney 2006). between identical twins. JAMA 1956; 160: 13. Ponticelli C, Yussim A, Cambi V et al. A ran- 277–282. domised, double-blind trial of basiliximab 3. Kuss R, Bourget P. An Illustrated History of Organ immunoprophylaxis plus triple therapy in kidney Transplantation. The Great Adventure of the transplant recipients. Transplantation 2001; 72: Century. Rueil-Malmaison: Sandoz, 1992: 45. 1261–1267. 4. Meiser-Kriesche HU, Schold ID, Srinivas TR et al. 14. Lawen JG, Davies EA, Mourad G et al. Lack of improvement in renal allograft survival Randomised double-blind study of immuno- despite a marked decrease in acute rejection rates prophylaxis with basilixmab, a chimeric anti- over the most recent era. Am J Transplant 2004; interleukin-2 receptor monoclonal antibody, in 4: 378–383. combination with mycophenolate mofetil- 5. Evans RW, Manninen DL, Garrison LP et al. The containing triple therapy in renal transplanta- quality of life of patients with end stage renal dis- tion. Transplantation 2003; 75: 37–43. ease. N Engl J Med 1985; 312: 553–559. 15. Webster AC, Playford EG, Higgind G et al. Interleukin 2 receptor antagonists for renal trans- 6. Aranzabal J, Perdigo L, Mijores J, Villar F. Renal plant recipients: a meta-analysis of randomised transplantation costs: an economic analysis and trials. Transplantation 2004; 77: 166–176. comparison with dialysis costs. Transplant Proc 16. Morris PJ, Russell NK. Alemtuzumab (Campath- 1991; 23: 2574. 1H): a systematic review in organ transplanta- 7. UK Transplant Activity. http://www.uktransplant. tion. Transplantation 2006; 81: 1361–1367. org.uk/ukt/statistics/statistics.jsp (accessed 12 17. Genberg H, Hansson A, Wernerson A et al. July 2006). Pharmacodynamics of rituximab in kidney allo- 8. Racusen LC, Solez K, Colvin RB et al. The Banff transplantation. Am J Transplant 2006; 6: 97 working classification of renal allograft 2418–2428. pathology. Kidney Int 1999; 55: 713–723. 18. Remuzzi G, Lesti M, Gotti E et al. Mycophenolate 9. Matas AJ. Acute rejection is a major risk factor for mofetil versus azathioprine for prevention of chronic rejection. Transplant Proc 1998; 30: acute rejection in renal transplantation (MYSS): a 1766–1768. randomised trial. Lancet 2004; 364: 503–512. 10. Pirsch JD, Ploeg RJ, Gange S et al. Determinants 19. Ojo AO, Meier-Kriesche H, Hanson JA et al. of graft survival after renal transplantation. Mycophenolate mofetil reduces late renal allo- Transplantation 1996; 61: 1581–1586. graft loss independent of acute rejection. 11. Agha IA, Rueda J, Alvarez A et al. Short course Transplantation 2000; 69: 2405–2409. induction immunosuppression with thymo- 20. Kramer BK, Montagnino G, Del Castillo D et al. globulin for renal transplant recipients. Efficacy and safety of tacrolimus compared with Transplantation 2002; 73: 473–475. cyclosporine A microemulsion in renal trans- References 125

plantation: 2 year follow-up results. Nephrol Dial Management of Cytomegalovirus Disease after Solid Transplant 2005; 20: 968–973. Organ Transplantation, 2nd edn. London: British 21. Margreiter R. European tacrolimus versus Transplantation Society, 2004. ciclosporin microemulsion renal transplantation 29. Lichtenstein IH, MacGregor RR. Mycobacterial study group. Efficacy and safety of tacrolimus infections in renal transplant recipients: report of compared with ciclosporin microemulsion in five cases and review of the literature. Rev Infect renal transplantation: a randomised multicentre Dis 1983; 5: 216–226. study. Lancet 2002; 359: 741–746. 30. Kasiske BL, Snyder JJ, Gilbertson DT, Wang C. 22. Wyeth. Summary of product characteristics for Cancer after kidney transplantation in the United Rapamune. http://emc.medicines.org.uk/emc/ States. Am J Transplant 2004; 4: 905. industry/default.asp?page=displaydoc.asp&doc 31. Information about skin cancer for patients with umentid=5747 (accessed 26 September 2006). an organ transplant. http://www.bad.org.uk/pub 23. Watson CJE, Firth J, Williams PF et al. A ran- lic/leaflets/transplant.asp (accessed 15 July 2006). domized controlled trial of late conversion from 32. Andreone P, Gramenzi A, Lorenzini S et al. Post- calcineurin inhibitor-based to sirolimus-based transplantation lymphoproliferative disorders. immunosuppression following renal transplanta- Arch Intern Med 2003; 163: 1997. tion. Am J Transplant 2005; 5: 2496–2503. 33. Paya CV, Fung JJ, Nalesnik MA et al. Epstein-Barr 24. Zimmerman JJ, Kahan BD. Pharmacokinetics of virus-induced posttransplant lymphoproliferative sirolimus in stable renal transplant patients after disorders. ASTS/ASTP EBV-PTLD Task Force and multiple oral dose administration. J Clin The Mayo Clinic Organized International Con- Pharmacol 1997: 37: 405–415. sensus Development Meeting. Transplantation 25. Baxter K, ed. Stockley’s Drug Interactions, 7th edn. 1999; 68: 1517. London: Pharmaceutical Press, 2006. 34. Choquet S, Leblond V, Herbrecht R et al. Efficacy 26. Vincenti F, Larsen C, Durrbach A et al. and safety of rituximab in B-cell post- Costimulation blockade with belatacept in transplantation lymphoproliferative disorders: renal transplantation. N Engl J Med 2005; 353: results of a prospective multicenter phase 2 study. 770–781. Blood 2006; 107: 3053. 27. Watschinger B, Pascual M. Capillary Cd4 deposi- 35. Schafer Keller P, Lyon S, Van Gelder F, De Geest tion as a marker of humoral immunity in renal S. A practical approach to promoting adherence allograft rejection. J Am Soc Nephrol 2002; 13: to immunosuppressive medication after renal 2420–2423. transplantation. Curr Opin Nephrol Hypertens 28. Newstead CG. Guidelines for the Prevention and 2006; 15 (Suppl 2): S1–S6.

10

Drug dosing in patients with renal impairment and during renal replacement therapy

Anne Millsop

The kidney plays an important role in the hand- reduced in patients with renal impairment as a ling of drugs in the body. Patients with renal result of: impairment may therefore require different • Nausea, vomiting or diarrhoea associated dosage regimens to patients with normal renal with uraemia3 function. Unfortunately, there are no absolute • Hypoproteinaemic oedema of the gastro- guidelines on how to adjust doses in renal intestinal tract (e.g. in nephrotic syndrome)3 impairment, and pharmaceutical company liter- • Reduced intestinal motility and gastric emp- ature often excludes patients with renal impair- tying time (e.g. in uraemic neuropathy)3, ment in the dosage guidelines.1 Even when which can reduce the time taken for the drug information can be found the advice may not to be absorbed but does not generally affect be specific and different texts may give differ- the extent of absorption4 ent advice.2 It is important, therefore, for • An increase in pH in the gut from increased pharmacists to have an understanding of the gastric ammonia production in uraemia, potential pharmacokinetic and pharmaco- which reduces the bioavailability of drugs dynamic changes in renal impairment so that requiring an acidic environment for appropriate dosing decisions can be made. absorption3,4 • Co-administration of drugs which increase

gastric pH (e.g. H2-antagonists) Drug handling considerations in • Co-administration of chelating agents such patients with renal impairment as those used for binding phosphate (see Chapter 6). Changes to the way drugs are handled in the It is also speculated that the absorption of presence of impaired kidney function will affect some drugs is increased as a result of: both the choice of drug and the method of • Reduced activity of drug-metabolising en- administration. Renal impairment can affect the zymes in the intestine, although this effect absorption, distribution, metabolism and elim- may be offset by increased first-pass metabol- ination of a drug. There may also be phar- ism in the liver4 mocodynamic changes. • Co-administration of drugs that increase gastric pH, which will increase the bio- availability of weakly acidic drugs.4 Absorption Drug doses are not routinely altered to allow Absorption of orally administered drugs may be for these factors alone but must be considered

127 128 Chapter 10 • Drug dosing in patients with renal impairment in the whole picture. If therapeutic levels of unbound, and therefore active, drug in the drugs are not being achieved or if a fast onset plasma. However, as there is more unbound of action is required, a change of dose or a drug available for metabolism this effect is usu- different route of administration may be ally transient. required. For highly bound drugs care must be taken in interpreting TDM results as total drug con- centrations (bound and unbound) are usually Distribution reported and not free active drug. In this instance low TDM levels may not necessarily be Changes to the distribution of drugs in the body subtherapeutic. The most important example of in patients with renal impairment may occur as this is phenytoin. Free phenytoin levels should a result of: be measured where possible. However, as free levels are not usually readily available it is • Changes in the hydration state of the patient important to know how to interpret a total • Alterations in protein binding plasma level. Equation 10.1 from Basic Clinical • Alterations in tissue binding. Pharmacokinetics6 may be used to adjust a total plasma level:

Changes in the hydration state of the patient Cp C = The state of hydration of a patient is only p Normal binding (1 – α) [P/P ] + α important for drugs with a small volume of dis- NL tribution (V ) (<50 L), such as gentamicin.5 In d where C is the plasma drug con- the presence of oedema the V will be increased p Normal binding d centration that would have been observed if the and in the presence of dehydration the V will d patient’s serum albumin concentration had be reduced. It is important to note the state of been normal; C is the observed plasma con- hydration when blood samples are taken for p centration reported by the laboratory; α is the therapeutic drug monitoring (TDM) and to use normal free fraction of phenytoin (0.1 for non- this knowledge when interpreting the results. renal failure patients and 0.2–0.35 for patients This is particularly important when the state of with renal failure, quoted as creatinine clear- hydration is fluctuating with the use of intra- ance <25 mL/min); P is the patient’s serum venous fluids, diuretics or intermittent renal albumin in units of g/dL; and P is the normal replacement therapy (RRT). NL serum albumin (4.4 g/dL). Where creatinine clearance is <10 mL/min Alterations in protein binding and the patient is undergoing haemodialysis treatment an equation incorporating a factor Protein binding is altered due to: which takes into account both altered serum • Hypoalbuminaemia albumin concentration and decreased binding • Uraemia and the accumulation of metabo- affinity for this patient group should be used: lites and endogenous substances, which will compete with the drugs for binding to albu- Cp min C = p Normal binding (0.48)(1 – α) [P/P ] + α • Altered structural arrangement of albumin NL possibly reducing the affinity or number of For this equation α has a value of 0.1.6 Although binding sites for drugs.3,4 these equations can help us determine TDM Alterations in protein binding are clinically results more accurately they are still only an important for highly protein-bound drugs estimation. Clinical signs and symptoms must (>80%).3 A reduction in bound drug in the also be used, and free levels of phenytoin plasma will result in a higher proportion of should be specifically requested. Assessment of renal function 129

Before taking samples for TDM consideration to which drugs are affected depends on the per- must be given to the time taken for equilibrium centage of active drug or active metabolite that to be reached (4 half-lives), as for some drugs would normally be excreted by this route. For the half-life is increased in renal impairment. some drugs accumulation of active metabolites with different properties to the active parent may change the pharmacological response in Alterations in tissue binding renal impairment. An example of this is peth- Alterations in tissue binding may affect the vol- idine. An excess of pethidine produces CNS ume of distribution (Vd) of a drug. For the depression, but an accumulation of the renally majority of drugs this is not clinically relevant.4 excreted metabolite norpethidine produces CNS One drug for which it is relevant is digoxin. The stimulation and seizures.7

Vd of digoxin may be reduced by up to 50% in In order to make decisions on doses of drugs established renal failure patients. Reduced load- excreted by the kidney an assessment of renal ing doses will need to be given to prevent tox- function needs to be made (see below). icity. Maintenance doses of digoxin are also markedly reduced because it is predominantly renally excreted. Pharmacodynamic alterations

Although there is not as much literature on Metabolism changes in the body’s response to drugs in renal impairment it is known that patients with Phase I and phase II metabolism is slower in uraemia have: chronic kidney disease.4,7 The effect of this is to • An increased sensitivity to drugs acting on increase serum drug concentrations of the par- the central nervous system such as benzo- ent drug. This may lead to higher prevalence of diazepines adverse effects and toxicity where drugs are • A reduced sensitivity to some endogenous usually metabolised to inactive metabolites. The hormones such as growth hormone kidney itself is also the site of metabolism for • An increased sensitivity to cholinesterase some drugs. Two important examples are the inhibitors hydroxylation of 25-hydroxycholecalciferol to • An increased risk of gastrointestinal bleeding active vitamin D (1α,25-dihydroxycholecalcif- with irritant drugs such as non-steroidal anti- erol) and the metabolism of insulin. For patients inflammatory drugs requiring vitamin D, either the active drug (cal- • An increased risk of hyperkalaemia with citriol) or a preparation requiring metabolism to drugs such as potassium-sparing diuretics.8 active drug by the liver (alfacalcidol) should be used. For diabetic patients insulin requirements will be reduced. Assessment of renal function Elimination Assessing the degree of renal impairment usu- The kidney is involved in the elimination of ally involves a measurement of glomerular fil- drugs and metabolites by glomerular filtration, tration rate (GFR). GFR cannot be measured renal tubular secretion and resorption.4 In renal directly so it is estimated from the clearance of impairment all these functions are reduced. The a solute present in a stable concentration in the reduction in glomerular filtration and tubular plasma and freely filtered by the kidney.9 The secretion results in higher plasma levels of drug solute ideally should not be secreted or re- and the reduction in resorption results in higher absorbed by the renal tubule. Naturally occur- concentrations of drug in the urine. The extent ring solutes which have been used are urea, 130 Chapter 10 • Drug dosing in patients with renal impairment creatinine and cystatin C. Exogenous products for GRF assessment. The ways creatinine clear- which have been used include inulin, isotopes ance (CrCl) by the kidneys can be estimated are and iohexol. The merits of each will be consid- given below. ered in turn.

Cystatin C Endogenous solutes Cystatin C is a low-molecular-weight protein that is produced at a constant rate and freely Urea filtered by the glomerulus.9 After filtration it is reabsorbed and catabolised by the tubular Urea is used with other results as a guide to the epithelial cells which means that it is only severity of renal impairment but its use to cal- present in the urine in small amounts. Plasma culate GFR is limited by external factors that levels of cystatin C correlate well with GFR and affect urea production. These include alterations may be more accurate than creatinine estima- in protein intake by the patient (which alters tions in mild renal impairment.10 Cystatin C production throughout the day); catabolism levels are not routinely used at present. caused by sepsis or treatment with cortico- steroids; hyperthyroidism; liver disease; gastro- intestinal bleeding and muscle injury. Another problem with using urea as a filtration marker is Exogenous solutes that it undergoes resorption by the renal tubules as well as being filtered, therefore it gives an Inulin underestimate of GRF and at low urine flow Inulin was considered the gold standard in rates its clearance is independent of GFR.9 measuring GFR.9 It is an inert polymer of fruc- tose that is freely filtered and is neither secreted Creatinine nor reabsorbed by the tubules. Measurement was by infusion of inulin to produce steady state Creatinine is a product of muscle breakdown plasma levels and an accurately timed urine formed by non-enzymatic degradation of collection. Inulin is no longer used as it is not muscle creatinine.9 Creatinine production is readily available in the UK and is difficult to relatively constant in any individual with a assay. stable diet, making it a good filtration marker. The production of creatinine varies between individuals as it is proportional to muscle mass Isotopes and meat intake. It is therefore important to take into account the age, sex, race and weight Radioactive chromium-51-ethylenediaminetetra- of each individual, and to ensure the indi- acetic acid (EDTA) can be injected and accurate vidual has a stable diet. Creatinine is actively blood samples taken following injection to estim- secreted in the proximal tubule as well as being ate GRF. The method measures disappearance filtered, so measuring creatinine clearance leads from the plasma rather than renal clearance but to an overestimate of GFR by 10–20 mL/min/ the results correlate well with inulin studies.9 1.73 m2.10 Creatinine secretion in the tubules is The disadvantage is that the patient is exposed blocked by some drugs (e.g. , tri- to radiation. Technetium-99-diethylenetriamine methoprim, probenecid, amiloride, spironolac- pentaacetic acid (DTPA) can also be used. GRF tone, triamterene). Administration of these can be measured by dynamic renal scanning to drugs in patients with renal impairment can measure the rate of disappearance of the isotope produce an increase in serum creatinine with- or by blood samples or collection of urine out altering the GFR. Despite these limitations, samples. Currently these assay methods are not creatinine is the most commonly used marker in routine use. Assessment of renal function 131

Iohexol This equation takes into consideration the differences in muscle mass (and therefore creat- Iohexol is a contrast medium that can be meas- inine production) between men and women ured with high-performance liquid chromato- and for different age groups. However it does graphy. It shows good correlation with inulin not take into account variations between differ- techniques as a filtration marker. Currently this ent races. It can only be used if the renal func- assay method is not in routine use. tion is stable, and it becomes inaccurate as the GFR falls because creatinine excretion is not solely by filtration (see above). It should not be Estimation of renal function used for patients in end stage renal failure or receiving RRT. Also the equation is reliant on Calculation of creatinine clearance accurate serum creatinine measurement and studies have shown that these results vary If a 24-hour urine collection has been carried between laboratories.11 Despite all of these out then a creatinine clearance can be calcu- points the Cockcroft and Gault equation is lated using the following equation:5 widely used because it can be quickly calculated at the patient’s bedside using readily available UV CrCl (mL/min) = laboratory results. S

Where U is the urine creatinine concentration Estimated GFR in µmol/L; V is the urine flow rate in mL/min The Modification of Diet in Renal Disease (i.e. volume of urine collected divided by the (MDRD) study group compared different time over which it was collected) and S is the methods for calculating GRF and found the serum creatinine concentration (sample taken following equation to give a good prediction midway through the 24-hour collection period) of GFR:12 in µmol/L. When conducted properly this is a very ac- –0.999 –0.176 eGFR = 170 (Pcr) (age) curate way of determining creatinine clearance, (0.762 if female) (1.180 if African however inaccuracies in collection often occur American) [SUN]–0.170 [Alb]+0.318 and it is cumbersome to perform. An alternative method is to calculate the 2 where eGFR is expressed in mL/min/1.73 m , Pcr creatinine clearance using serum creatinine is plasma creatinine concentration in mg/dL, levels. The most widely used calculation for this age is in years, SUN is serum urea nitrogen con- is the Cockcroft and Gault equation: centration in mg/dL and Alb is serum albumin concentration in g/dL. The equation was devel- F (140 – age [years]) oped in patients with chronic kidney disease, CrCl [mL/min] = ideal body weight [kg] who were predominantly white and did not Serum creatinine [µmol/L] have diabetic kidney disease or a kidney trans- plant. The equation has been validated for where F is 1.23 for men and 1.04 for women. African Americans but no other ethnic groups. The ideal body weight is used for patients The authors report that this method is more who are obese and is calculated as follows: accurate at estimating GFR than the Cockcroft and Gault equation.12 However the equation Ideal body 50 kg + 2.3 kg for every inch assumes that all patients have a body surface = 2 weight (men) over 5 feet in height area (BSA) of 1.73 m . For patients with a BSA less than 1.73 m2 the eGFR is likely to over- Ideal body 45.5 kg + 2.3 kg for every estimate their kidney function and for patients = weight (women) inch over 5 feet in height. with a BSA greater than 1.73 m2 the eGFR is 132 Chapter 10 • Drug dosing in patients with renal impairment likely to underestimate their kidney function. order to limit the potential problems with Use of the patient’s actual BSA will overcome adverse effects and toxicity, as far as possible this problem, but this requires the patient’s drugs with the following characteristics should height and weight to be available. Simplified be used: versions of the equation using four rather than six variables are also in use.10 In units where the • Not nephrotoxic MDRD prediction equation is being used the • Has a large therapeutic index eGFR is calculated automatically by the report- • Does not require renal metabolism to an ing laboratory. active form When considering drug doses it is important • Does not require renal excretion of active to remember that most standard texts use creat- drug or active metabolites inine clearance as an estimation of renal func- • Has a low adverse effect profile tion and not eGFR.13 For most drugs there is a • Is not highly protein bound broad range for guidance on dosage and so in • Has an action unaffected by altered tissue practice the variations of measurement will sensitivity not change the recommendations,14 but it is • Is unaffected by fluid balance changes important to consider the implications of • Is able to reach the site of action in high under- and overdosing for patients whose cal- enough concentrations in the presence of culated GFR varies according to which formula renal impairment. has been used. In addition, if a drug is to be given by the intravenous route it is preferable to use a drug with a low sodium content and requiring a Classification of renal function small infusion volume. It is rarely possible to find a drug fitting all Some texts giving advice on drug doses in renal these criteria, but where a drug has potential impairment classify renal impairment in terms renal adverse effects, serious dose-related of creatinine clearance values and some texts adverse effects, or a narrow therapeutic range use qualitative measurements such as ‘mild’, with no potential to monitor, an alternative 2 ‘moderate’ or ‘severe’ renal impairment. The drug should be sought. classification for each literature source should Once a drug has been selected, a decision on always be followed but dosage decisions are the dose needs to be made. Ideally one or more more difficult when quantitative values are not of the available renal drug dosing reference given. sources should be consulted along with the summary of product characteristics (SPC), and advice sought from a renal pharmacist who may Adjusting doses for patients with have clinical experience of using the drug. renal impairment When interpreting the dose recommendations, remember that eGFR and creatinine clearance values are not interchangeable (see Calculation It can be seen from the information above that of renal function above). The following prin- assessing the degree of renal impairment is not ciples should also be applied: an exact science. The same is applicable to the dosing of these patients. There are many literat- • If the drug is unaffected by renal impairment ure sources available with suggested doses for it may be used in usual doses and the patient patients with renal impairment (see Further should be monitored for signs of increased reading) but the advice in different sources is sensitivity to the effects of the drug or to the not always consistent.2 It is important therefore adverse effects. to be able to apply your pharmaceutical knowl- • Drugs that require therapeutic levels quickly edge to each individual case. may require a loading dose as the time taken The first step is the choice of drug to use. In to reach steady state will be prolonged for Drug handling in renal replacement therapy 133

drugs where the metabolism and excretion is Drug handling considerations in slowed in renal impairment. patients undergoing renal • Maintenance doses and frequency will replacement therapy depend on the extent of renal impairment and the drug to be used. Once a patient requires RRT their own kidney function is negligible and so removal from the Loading doses body of a drug dependent on renal excretion is also negligible. It is therefore important to know The following equation can be used to calculate whether a drug is likely to be removed by RRT. a loading dose: The factors to consider during RRT fall into two categories: Loading Target plasma concentration • The drug characteristics that will affect drug dose (mg) = (mg/L) Volume of removal distribution (L/kg) • The RRT characteristics that will affect drug Ideal body weight (kg) removal.

There will be variations in the loading dose cal- culated depending on the target concentration Drug characteristics affecting drug removal chosen (most drugs have a target range) and in RRT depending on the volume of distribution figure used (this can vary between literature sources The following drug characteristics are likely to and may again be quoted as a range). Knowl- affect drug removal by RRT: edge of the adverse effect and toxicity profile • Molecular weight will be important in choosing the loading dose. • Percentage of drug bound to plasma proteins • Volume of distribution Maintenance doses • Water solubility • Percentage of renal clearance in normal sub- The following equation can be used to calculate jects maintenance doses:5 • Steric hindrance. Knowledge of the molecular weight is more DRrf = DRn [(1 – Feu) + (Feu RF)] important for drug removal by intermittent haemodialysis than for continuous haemofiltra- where DRrf is the dosing rate in renal failure, tion or haemodiafiltration. For haemodialysis, DRn is the normal dosing rate, RF is the extent molecules with a molecular weight >500 Da are of renal failure (calculated as patient’s CrCl not likely to be removed, whereas for haemo- (mL/min) divided by the ideal CrCl of 120 filtration using a highly permeable membrane mL/min) and Feu is the fraction of drug norm- molecules with a molecular weight of up to ally excreted unchanged in the urine (available 30 000 Da may be removed.15,16 As only in literature sources and the SPC). unbound drug will be available for removal by Once a dosing regimen has been decided it is RRT, knowledge of the percentage of drug that important to monitor the patient’s progress. For is protein bound is important. Drugs with low drugs with a narrow therapeutic range serum protein binding potential are more likely to be drug levels should be monitored and the removed. It is also important to remember that physiological response to the drug should also changes in protein binding occur in patients be monitored. The patient and his or her carers with low serum albumin levels and uraemia as should be informed of potential adverse effects discussed above. The volume of distribution of or signs of toxicity so that they can report any each drug is important as only free drug in the they may experience. plasma is available for removal by RRT. Drugs 134 Chapter 10 • Drug dosing in patients with renal impairment

with a Vd < 1 L/kg are more likely to be removed by dialysis.17 Care must be taken in interpreting Table 10.1 System characteristics important for drug literature on Vd as alterations in protein bind- removal by haemodialysis, continuous peritoneal ing in renal impairment will alter the volume of dialysis or continuous haemofiltration distribution of highly protein-bound drugs (see System characteristics important above). For intermittent haemodialysis only for drug removal small fractions of drugs with a high Vd will be removed. For continuous RRT, drugs with a high Haemodialysis Duration of the dialysis procedure Vd will be more effectively removed because the Blood flow rate in the dialyser equilibrium between the plasma and tissue Dialysate flow rate levels will be constantly changing. The water Composition of the dialysate solubility of a drug will reflect the level of drug Type of membrane used usually in the plasma and therefore available for Peritoneal dialysis Volume of exchange of dialysate removal by RRT. The higher the water solubility Frequency of exchange of the more likely the drug is to be removed.17 dialysate Another reflection of a drug’s likelihood of Composition of the dialysate being removed is the amount of drug usually Osmotic gradient between removed by the kidneys. Although RRT cannot dialysate and plasma mimic completely the role of the kidneys, it is Pathology of the peritoneum more likely to remove drugs usually filtered by Continuous Type of membrane used the kidney in normal subjects. Lastly steric haemofiltration Selection of A-V or V-V system used Pump pressure (if used) hindrance is important. Even if a molecule appears small enough to be removed by the RRT process the atomic arrangement of the drug molecule will affect its ability to pass through the RRT membrane. For continuous peritoneal dialysis the mem- brane used is the peritoneal membrane and the health of this membrane is important for drug RRT system characteristics affecting drug removal. During peritonitis the membrane removal becomes more porous and will remove a larger proportion of drugs and solutes. Larger volumes The system characteristics affecting drug of exchange and frequent changes will increase removal will vary depending on the type of RRT the concentration gradient and aid diffusion of used. The characteristics important to each sys- drugs out of the plasma. The composition of the tem are listed in Table 10.1. dialysate used will also affect the rate of diffu- For intermittent haemodialysis, the duration sion. Lastly the osmotic gradient is important, a of dialysis will affect drug removal. For example, high gradient will remove more water and more drug will be removed in a 4-hour session therefore more water-soluble drugs with it. than a 3-hour session. The flow rate of both The most important factors influencing drug the blood and dialysate is important. Drug removal in continuous haemofiltration are the removal by haemodialysis is predominantly by membrane permeability and the system pressure diffusion, increasing the flow rate will therefore (which depends on whether an arteriovenous or result in a higher concentration gradient venovenous system is used and the pump pres- between the two and more drug being removed. sure, see Chapter 8). This is because convective The composition of the dialysate will also transport and ultrafiltration are the predomin- affect the diffusion process. Lastly, there are dif- ant factors for drug removal in haemofiltration. ferent membranes available for dialysis, the type For continuous haemodiafiltration a combina- of membrane will affect drug removal because tion of the haemodialysis and continuous of changes in permeability and composition haemofiltration factors will apply. For any sys- which favour removal of different drugs.16 tem using a membrane, interactions between Adjusting drug doses for renal replacement therapy 135 the drug and membrane may occur. Adsorption therapy not involving the use of a pumped onto the synthetic polymer membrane will circuit (CAVHF, CAVHDF) will allow drug doses reduce the drug clearance. Adsorption is for a clearance in the range of 10–20 mL/min dependent on blood pH, drug and membrane. to be used. However, those utilising pumped These are complex interactions and cannot systems (i.e. CVVHF and CVVHDF) have an always be predicted. even better solute clearance, and in the case of CVVHDF, clearances can be in the range of 30–40 mL/min, allowing for much larger drug doses to be prescribed. Adjusting drug doses for patients Dosage decisions can also be made using a undergoing renal replacement calculation involving the sieving coefficient of therapy the drug. The sieving coefficient is a measure of the proportion of drug that will pass For patients receiving RRT a creatinine clearance through the haemofiltration membrane during cannot be calculated using the equations dis- ultrafiltration.3 A drug which passes freely cussed above. Table 10.2 gives a guide to the through the membrane will have a sieving clearance provided by each system. coefficient of 1 and a drug which does not pass However, it is important to know the type of through the membrane will have a sieving dialysis and membrane used as clearances will coefficient of 0. A knowledge of the machine vary as described above. The manufacturer of clearance and the sieving coefficient can be the machine and filters should be able to give combined to calculate the drug clearance for an estimate of creatinine clearance by their sys- that system: tem. Haemodialysis is more effective than peri- toneal dialysis at removing fluid and solutes. Machine clearance Drug clearance = However, because the procedure is intermittent Sieving coefficient (often every 2–3 days) drugs should be dosed as if for a creatinine clearance of <10 mL/min The sieving coefficient can be found in spe- unless the SPC or other specialist texts state cialist texts but is dependent on the type of differently. If a drug is likely to be removed by haemofiltration and membrane used. Once an dialysis it should be given after the procedure idea of the rate of drug clearance in RRT has and not before. For continuous peritoneal dia- been established, literature sources such as renal lysis doses for creatinine clearance <10 mL/min drug dosing textbooks and the SPC may be con- are also used. Continuous renal replacement sulted to find information on dosing.

Table 10.2 Creatinine clearance for different systems of renal replacement therapy

Renal replacement therapy Typical theoretical GFR achieved during therapy (mL/min)

Intermittent haemodialysis 150–200 during dialysis (0–10 between dialysis periods) Continuous arterio-venous haemofiltration (CAVHF) 0–15 Continuous veno-venous haemofiltration (CVVHF) 15–25 Continuous arterio-venous haemodiafiltration (CAVHDF) 20 Continuous veno-venous haemodiafiltration (CVVHDF) 30–40 Continuous ambulatory peritoneal dialysis (CAPD) (4 exchanges daily) 5–10

Data from Industry Submission, Renal National Service Framework (Acute Renal Failure), October 2003.17 136 Chapter 10 • Drug dosing in patients with renal impairment

Loading doses Summary

For drugs requiring therapeutic levels quickly Drug dosing in patients with impaired renal and with a prolonged half-life in renal failure a function is a complex area. It is important to loading dose may be required. The equation have an understanding of how drug handling above can be used to calculate the loading dose. may be altered in renal impairment and RRT and on the limitations of the calculations used to estimate renal function in order to help make Supplementary doses for intermittent renal informed decisions on drug doses. Where poss- replacement therapy ible, textbooks on drug dosing in renal impair- ment should be consulted and also make use of Some texts quote supplementary doses to be the pharmacists with a special interest in the given after intermittent RRT. These will only be field who have clinical experience with these important for drugs with a low V and a narrow d patients. In all cases once a drug regimen has therapeutic range. It is better to adjust the tim- been prescribed monitor the patient for efficacy, ings of the regular doses so that the next dose adverse effects and signs of toxicity. falls after the RRT session rather than add in extra doses. C CASE STUDY

Mr PT is a haemodialysis patient. He dialyses three times a week via a tunnelled line. The haemodialysis registrar has admitted him to the renal ward suffering with rigors and fever (39ºC) and has prescribed him IV flucloxacillin.

Current medication: • Flucloxacillin IV 1 g four times daily • Alfacalcidol 0.25 µg three times weekly • Calcium acetate 2 tablets with meals • Atenolol 50 mg daily • Lisinopril 5 mg daily • Erythropoietin 2000 units SC three times weekly • Iron sucrose 100 mg IV weekly.

Blood results pre-dialysis on the day of admission: • Creatinine 505 µmol/L (70–120 µmol/L) • Urea 26 mmol/L (3.7–8.4 mmol/L) • Sodium 138 mmol/L (134–148 mmol/L) • Potassium 5.5 mmol/L (3.5–5 mmol/L).

Q1. Is the dose of flucloxacillin reasonable for a haemodialysis patient?

Q2. Should supplementary doses be given on dialysis days?

Q3. How should Mr PT be monitored? Further reading 137

References 12. Levey AS, Bosch JP, Lewis JB et al. A more accu- rate method to estimate glomerular filtration rate from serum creatinine: a new prediction equa- 1. Martin-Facklam M, Rengelshausen J, Tayrouz Y et tion. Modification of Diet in Renal Disease Study al. Dose individualisation in patients with renal Group. Ann Intern Med 1999; 130: 461–470. insufficiency: does drug labelling support optimal 13. Bauer L. Creatinine clearance versus glomerular management? Eur J Clin Pharmacol 2005; 60: filtration rate for the use of renal drug dosing in 807–811. patients with kidney dysfunction. Pharmaco- 2. Vidal L, Shavit M, Fraser A et al. Systematic com- therapy 2005; 25: 1286–1287. parison of four sources of drug information 14. Thompson CA. Better renal-function estimates regarding adjustment of dose for renal function. not expected to alter drug dosing right away. Am BMJ 2005; 331: 263–266. J Health-Syst Pharm 2005; 62: 2442–2444. 3. Aweeka F T. Dosing of drugs in renal failure. In: 15. Davies JG, Kingswood JC, Street MK. Drug Koda-Kimble MA, Young LY, eds. Applied removal in continuous haemofiltration and Therapeutics: The Clinical Use of Drugs, 7th edn. haemodialysis. Br J Hosp Pharm 1995; 54: Philadelphia: Lippincott Williams & Wilkins, 524–528. 2001: 32.1–32.21. 16. Giles L. Renal replacement therapy. In: Elliott R, 4. Gabardi S, Abramson S. Drug dosing in chronic ed. Critical Care Therapeutics. London: Pharma- kidney disease. Med Clin N Am 2005; 89: ceutical Press, 1999: 49–56. 649–687. 17. Industry Submission, Renal National Service 5. Harper A. Acute renal failure. In: Walker R, Framework (Acute Renal Failure) October 2003. Edwards C, eds. Clinical Pharmacy and Thera- 18. Ashley C, Currie A, eds. Renal Drug Handbook, peutics, 2nd edn. London: Churchill Livingstone, 2nd edn. Oxford: Radcliffe Medical Press, 2004. 1999: 215–229. 6. Winter ME. Basic Clinical Pharmacokinetics, 3rd edn. Vancouver: Applied Therapeutics Inc., 1994. 7. Matzke GR, Frye RF. Drug administration in Further reading patients with renal insufficiency: Minimising renal and extrarenal toxicity. Drug Safety 1997; 16: 205–231. Ashley C, Currie A, eds. Renal Drug Handbook, 2nd 8. Aronson JK. Drugs and renal insufficiency. edn. Oxford: Radcliffe Medical Press, 2004. Medicine 2003; 31: 103–109. Bennett WM, Arnoff GR, Berns JS et al. Drug Prescribing 9. Mole DR, Mason PD. Assessment of renal func- in Renal Failure: Dosing Guidelines for Adults, 4th tion. Medicine 2003; 31: 5–10. edn. Philadelphia: American College of Physicians, 10. Stevens LA, Levey AS. Measurement of kidney 1999. function. Med Clin N Am 2005; 89: 457–473. Dollery C. Therapeutic Drugs, 2nd edn. London: 11. Schneider V, Henschel V, Tadjalli-Mehr K et al. Churchill Livingstone, 1999. Impact of serum creatinine measurement error Sweetman, SC ed. Martindale: The Complete Drug on dose adjustment in renal failure. Clin Reference, 35th edn. London: Pharmaceutical Press, Pharmacol Ther 2003; 74: 458–467. 2007.

11

Drug-induced kidney disease

Jane Pearson

Acute renal failure (ARF) is generally defined as autoregulation (Figure 11.1).3 Understanding a sustained rise in serum creatinine of at least these mechanisms makes it easier to guess 50 µmol/L, or greater than 20% rise above base- which type of drugs of may be responsible for line (if the baseline is 250 µmol/L), that occurs pre-renal ARF. over a period of days to several weeks. It is usu- Drugs that decrease renal perfusion will have ally accompanied by a fall in urine output. This an adverse effect on renal function. This is seen may present as anuria or oliguria. Anuria is a as an acute reduction in glomerular filtration complete lack of urine output and oliguria is rate (GFR), which is usually reversible on low urine output of less than 15–20 mL/h or about 500 mL in a normal adult. ARF is common, but the incidence depends Stimulation of juxtaglomerular apparatus on the definition being used. It is quoted in the literature as being anything between 72 to 620 cases per million. As many as 30% of all cases Renin release in presence of low circulating volume and pressure of ARF are thought to be secondary to drugs and 2–5% of hospital inpatients develop drug- induced renal failure.1 Activation of renin-angiotensin-aldosterone cascade Drug-induced ARF can occur with or without oliguria and different mechanisms are used to determine the patterns of drug-induced renal Angiotensin II mediated constriction of the post glomerular failure. ARF is often described as pre-renal, arteriole renal or post-renal, and drugs can be responsible for each of these types of kidney disease.2 Maintenance of glomerular filtration pressure

Pre-renal (or haemodynamic) acute renal failure Cyclo-oxygenase (COX) responsible for synthesis of a number of vasoconstrictive arachidonic acid metabolites, including

prostacyclin PG-I2 In healthy people, autoregulation maintains adequate renal blood flow as blood pressure changes. This homeostatic mechanism serves to Facilitate renin secretion, vasodilation of the pre-glomerular protect normal renal blood flow in a number of arteriole and regulation of different disease states that are characterised by sodium excretion hypotension (e.g. congestive heart failure and artherosclerotic renal artery stenosis). There are Figure 11.1 Autoregulation of renal blood flow through a number of hormones and autocrine factors hormones and autocrine factors acting on renal blood acting on renal blood vessels to mediate this vessels.

139 140 Chapter 11 • Drug-induced kidney disease restoring blood supply to the kidneys. When polycystic kidney disease and hypertensive not corrected, persistent renal hypoperfusion nephropathy. will ultimately lead to ischaemic acute tubular Angiotensin II has a number of intra-renal necrosis. Any drug that compromises circulation effects, including: may induce ARF. This can include drugs that • Regulation of renal blood flow decrease cardiac output and those that increase • Regulation of GFR peripheral vasoconstriction. • Regulation of tubular reabsorption of sodium • Inhibition of renin release. Volume depletion In patients with severe renal impairment or renal artery stenosis, angiotensin II acts within Volume depletion with water and electrolyte the kidney to constrict the efferent glomerular loss can occur with excessive use of laxatives or arteriole and maintains glomerular filtration. diuretic therapy, in particular loop diuretics. ACE inhibitors block this effect and cause This can cause particular problems in the vasodilatation of the efferent arteriole, leading elderly, but severe renal failure rarely occurs to a sudden decrease in GFR and hence ARF. except in patients with pre-existing renal dis- The frequency of ACE inhibitor-induced ease. Lithium in combination with diuretics renal failure varies between 6 and 23% in leads to synergistic toxicity. Non-steroidal anti- patients with bilateral stenosis and increases to inflammatory drugs (NSAIDs) can exacerbate 38% in patients with unilateral stenosis on a pre-renal effects by further decreasing renal per- single kidney. fusion. They impair the ability of the renal NSAIDs inhibit the production of vasculature to adapt to falls in perfusion pres- prostaglandins E2, D2 and I2 within the kidney. sure or to an increase in the vasoconstrictor bal- These are potent vasodilators that are crucial in ance. Hypokalaemia from laxative-induced maintaining renal circulation to the glomerulus diarrhoea or overuse of diuretics can produce a and medulla. This is especially important when chronic nephropathy. vasoconstrictor substances such as angiotensin II or antidiuretic hormone (ADH) are increased. This can occur in conditions such as heart fail- Altered renal haemodynamics ure and severe hypertension. Inhibition of prostaglandin synthesis leads to vasoconstric- Some drugs have specific effects on renal per- tion and a subsequent decrease in renal blood fusion. Haemodynamic ARF is a recognised flow, GFR and urine volume. complication of treatment with angiotensin- Renal function usually recovers if NSAID converting enzyme (ACE) inhibitors and therapy is withdrawn early enough, although angiotensin II receptor antagonists. These alter permanent damage can occur. There is little renal haemodynamics through their vasodilator evidence that NSAIDs impair renal function in effects on the efferent glomerular arterioles. otherwise healthy individuals. This is particularly evident in patients with pre- existing renal artery stenosis. A sharp rise in a patient’s serum creatinine after starting an Other ACE inhibitor may indicate the presence of bilateral renal artery stenosis. ACE inhibitor Some renally toxic drugs affect renal function use in such patients is contraindicated. Reno- by more than one mechanism, including vaso- vascular disease has been found in 34% of constriction. Renal transplant units have elderly patients with heart failure. Any renal immunosuppressive regimens based on using damage that has occurred is usually reversible if calcineurin inhibitors (ciclosporin or tacro- the drug is discontinued promptly. Other risk limus). High blood levels of ciclosporin and factors for ACE inhibitor-induced renal disease tacrolimus are associated with a negative effect are hypovolaemia, severe chronic heart failure, on renal haemodynamics. They cause intense Intrinsic renal toxicity 141 vasoconstriction of the microvasculature within Glomerular the kidney, resulting in reduced renal perfusion, a fall in GFR and hypertension. Drug-induced glomerulonephritis is an Doses must be adjusted to maximise the immune-mediated disease where antigen– therapeutic effect and minimise the renal dam- antibody complexes accumulate within the age. High blood levels will obviously be associ- glomerulus. You then get an inflammatory ated with renal damage, but low blood levels are response due to the depositing of immuno- associated with increased risk of rejection of the globulins and complement in the base mem- transplanted kidney. Many drugs that affect branes and blood vessels. The result is a reduced the cytochrome P450 system will interact with GFR, salt and water retention, increased these drugs to alter blood levels. intravascular volume and hypertension. Pro- teinuria may also be present and the patient can develop nephrotic syndrome. Many drugs are known to cause glomerulonephritis (Table 11.1). Intrinsic renal toxicity

This type of renal failure is often described as a Tubular ‘renal’ cause of renal failure, and can be sub- divided into four categories: vascular, glomer- Acute tubular necrosis (ATN) can occur due to ular, tubular and interstitial. renal ischaemia or nephrotoxic agents or both. It can arise from the direct toxic affect of drugs or their metabolites on the renal tubules. ATN Vascular can occur with normal doses, but more often results from high-dose treatment or accumula- The lumen of the blood vessels may become tion of the drug due to pre-existing renal blocked either by atheroembolic disease or by impairment. ARF may develop after short- or foreign material that causes an inflammatory long-term drug exposure. Risk factors include reaction that obliterates the lumen. This vas- advanced age, pre-existing renal impairment, culitis causes inflammation and necrosis in the hypertension, heart disease, peripheral vascular vessel wall. Drugs are not usually the cause of disease and diabetes. Drug-induced ATN is usu- vascular renal damage. ally associated with oliguria and a significant proportion of patients require renal replacement therapy on a temporary basis. Nephrotoxic agents should be avoided if at all possible in patients at highest risk of devel- oping ATN. Drugs that are most often associated with ATN are listed in Table 11.2. Table 11.1 Drugs known to cause glomerulo- Maintenance of adequate hydration and nephritis therapeutic drug monitoring where appropriate may minimise the risk of developing ATN. • Allopurinol • Gold • Dapsone • Hydralazine • Halothane • Penicillamine Interstitial • NSAIDs • Phenindione Acute interstitial nephritis (AIN) is a hyper- • Penicillins • Rifampicin sensitivity reaction that is characterised by a • Probenecid • Thiazide diuretics fall in GFR within hours, days or months of • Sulfonamides • Procainamide exposure to a particular drug. The decrease in • Tolbutamide • Psoralen • Captopril • Levamisole GFR is often associated with proteinuria and haematuria. The interstitium is infiltrated with 142 Chapter 11 • Drug-induced kidney disease

of the offending drug, but permanent impair- Table 11.2 Drugs associated with acute tubular ment may occur. necrosis Chronic interstitial nephritis is also known as analgesic nephropathy. It is often under- • Aciclovir • Furosemide diagnosed as patients under-report their use of • Aminoglycosides • Gold analgesics. It can cause acute and chronic renal • Amphotericin • Ifosfamide failure. ARF may result from allergic reactions, • Cephalosporins • Lithium direct toxic tubule damage or reduction on 4 • Cisplatin • Mannitol renal blood flow. Chronic damage is often seen • Contrast media • NSAIDs after prolonged use of analgesics. The analgesic • Ciclosporin • Paracetamol combinations containing salicylates, caffeine or • Ethylene glycol • Tacrolimus paracetamol seem to increase the risk of chronic • Foscarnet • Vancomycin tubular interstitial disease.

Post-renal damage (obstructive inflammatory cells, including eosinophils. The uropathy) other classic clinical signs are low-grade pyrexia, rash and arthralgia. A large number of different drugs are com- Obstruction to urine flow through the kidneys monly implicated in causing AIN (Table 11.3). can be caused by a number of factors. Ureteric AIN is thought to account for up to 15% of fibrosis, renal calculi, blood clots and mechan- hospital admissions due to ARF. It is important ical blockage can all occur with drug therapy. to identify the causative agent and withdraw Anticoagulants can cause bleeding in this drug from treatment in the patient. patients, which can subsequently lead to blood Recovery of renal function usually occurs over a clot formation and ureteric obstruction. period of one month to a year after withdrawal Retroperitoneal haemorrhage following overtreatment with anticoagulants can also cause ureteric obstruction due to external com- pression of the ureters. Patients with severe renal impairment may be at greater risk of this due to uraemia induced platelet dysfunction, Table 11.3 Drugs commonly implicated in causing leading to a greater risk of bleeding. acute interstitial nephritis Retroperitoneal fibrosis is overgrowth of fibrous tissue in the peritoneum. The ureters can • Allopurinol • Isoniazid become embedded in the fibrous tissue, causing • Aminosalicylates • Lithium unilateral or bilateral obstruction and renal • Amlodipine • Mesalazine failure. This has been associated with metho- • Azathioprine • NSAIDs trexate, methysergide, beta-blockers, methyl- • Bumetanide • Penicillins dopa, bromocriptine and pergolide. Ureteric • Carbamazepine • Phenobarbital fibrosis has been associated with analgesic • Cephalosporins • Phenytoin nephropathy. Symptoms of retroperitoneal • Cimetidine • Proton pump inhibitors fibrosis include malaise, back and flank pain • Co-trimoxazole • Quinolones and weight loss. Although the condition usually • Diltiazem • Ranitidine regresses on discontinuation of the causative • Erythromycin • Rifampicin drug, this tends to be slow and surgical inter- • Furosemide • Sulfonamides vention is often necessary to prevent further • Gentamicin • Thiazides deterioration in renal function. • Gold • Vancomycin Crystallisation of urate and stone formation • Interferon within the ureteric lumen predisposes to Conclusion 143 obstructive nephropathy. Treatment of myelo- Conclusion proliferative disorders with cytotoxic drugs can cause tumour lysis to occur. This can lead to uric acid crystals being deposited in the renal There are many factors involved in drug- tubules to such an extent that blockage occurs. induced renal failure, with some drugs having This is particularly a problem if the patient has more than one mechanism of causing damage. a high tumour burden. The use of prophylactic ARF is common in hospitalised patients and is allopurinol or rasburicase plus adequate fluid associated with significant morbidity and mor- intake may decrease this risk. tality. Pharmacists can play an important role in Crystalluria can also occur with sulfona- the management of this patient group. They can mides, acetazolamide, mercaptopurine, metho- identify possible drug causes of renal failure, trexate, cisplatin, probenecid, naftidofuryl, ensure further nephrotoxic drugs are not pre- aciclovir, indinavir, cidofovir and ganciclovir. A scribed during the recovery and treatment of high intake of fluid should be maintained in renal failure and advise on appropriate dose patients to minimise the risk of crystalluria. adjustments for drugs according to renal func- tion or renal replacement therapy.

C CASE STUDY

Mr NT is a 40-year-old father of two, diagnosed with testicular teratoma. He has normal renal function (EDTA-measured creatinine clearance of 92 mL/min) and is treated with: prednisolone, vincristine 1 mg/m2, methotrexate 300 mg/m2, bleomycin 7.5 mg 2 doses, cisplatin 120 mg/m2. Mr NT is 5 feet 6 inches tall and weighs 101 kg. His body surface area is 2.09 m2. One week later, a routine set of blood tests shows: • Urea 15.1 mmol/L (3.0–6.5 mmol/L) • Creatinine 200 µmol/L (60–120 µmol/L) • Sodium 137 mmol/L (135–145 mmol/L) • Potassium 2.8 mmol/L (3.5–5.0 mmol/L) • Magnesium 0.35 mmol/L (0.7–1.0 mmol/L) • Calcium (corrected) 1.87 mmol/L (2.2–2.65 mmol/L).

Q1. What are the metabolic abnormalities?

Q2. What has caused these abnormalities?

Q3. How can you avoid this complication? On his next admission, Mr NT was given his chemotherapy as per protocol. Two days later, he was found to be pyrexial with a temperature of 39.80ºC, and his full blood count revealed that he was neutropenic, with a WBC count of 1.3 109/L (normal range 3.7–11.0 109/L), and an absolute neutrophil count of 0.1 109/L (normal range 1.5–7.5 109/L).

(continued overleaf) 144 Chapter 11 • Drug-induced kidney disease

C CASE STUDY (continued)

He was prescribed empirically the following drugs: • Amikacin IV 700 mg twice daily • Ceftazidime IV 2 g three times daily • Metronidazole IV 500 mg three times daily. His biochemistry over the following days was as follows:

Day 2 Day 3 Day 4 (Normal range)

Sodium (mmol/L) 143 141 144 135–145 Potassium (mmol/L) 5.3 5.5 6.2 3.5–5.0 Calcium (mmol/L) 2.01 1.97 1.93 2.1–2.6 Magnesium (mmol/L) 0.97 0.86 0.73 0.7–1.0 Phosphate (mmol/L) 1.29 1.57 2.01 0.70–1.25 Urea (mmol/L) 7.3 10.6 15.4 3.0–6.5 Creatinine (µmol/L) 155 235 481 60–120

Q4. What has happened now?

Q5. Comment on the antibiotic therapy he has been prescribed. Do you need to intervene?

Q6. What would you recommend?

References induced renal failure. Prescriber 5 Feb 2006: 59– 63. 3. Ashley C. Renal failure – how drugs can damage 1. Ashley C, Holt S. Renal disease (1): acute renal fail- the kidney. Hosp Pharm 2004; 11: 48–53. ure. Pharm J 2001; 266: 625–628. 4. Lameire N, Van Biesen W, Vanholder R. Acute renal 2. Waring S. Features and management of drug- failure. Lancet 2005; 365: 417–430. 12

Autoimmune kidney disease

Marc Vincent

Autoimmunity is an abnormal condition in antibody formation is often not known or is which the body reacts with constituents of its poorly understood. own tissues. The normal response of the Type II reactions describe cytotoxic reactions immune system is to produce antibodies to for- resulting when an antibody reacts with the eign antigens such as bacteria or viruses. antigenic components of a cell or tissue. The Activated B-cells recognise the foreign antigen antigen–antibody formation may activate cyto- and stimulate antibody production. This reac- toxic cells such as killer T-cells or macrophages tion activates the complement system, which which cause cell injury and usually involves amplifies the B-cell response causing lysis of complement activation. This antibody-mediated antigenic cells. Autoimmunity occurs when the reaction is seen in Goodpasture’s syndrome immune system produces antibodies to self- secondary to antiglomerular basement mem- antigens in body tissues (autoantibodies). brane antibodies (anti-GBM) and in systemic Autoimmune kidney damage usually occurs as a vasculitis secondary to antineutrophil cyto- result of autoantibodies directed towards the plasmic antibodies (ANCA). antigens in the glomerulus, causing glomer- Type III reactions involve deposition of ulonephritis. antibody–antigen immune complexes in vessels The more common autoimmune kidney dis- or tissues. The immune complexes may be de- eases are shown in Figure 12.1 and it is these posited in various tissues where they activate which will be discussed here. There are other the complement system, causing the release of kidney diseases which may come under the lysosomal enzymes and cytokines and cell umbrella of autoimmune kidney disease but injury. Kidney damage results when im- they are less common and beyond the scope of mune complexes localise in the glomerulus. this chapter. Immune complexes are deposited in the The clinical manifestations associated with glomerular capillary wall, causing an inflam- autoimmune kidney disease can be disease- matory response and glomerulonephritis. specific but those associated with glomerular Several autoimmune kidney diseases secondary injury are often the same or similar. Definitive to immune complex formation are shown in diagnosis is based on immunological determina- Figure 12.1. tion and kidney biopsy. The immunological markers of the different diseases are shown in Figure 12.1.1 Clinical manifestations

Regardless of the pathogenesis, patients usually Pathogenesis present with one of six clinical syndromes: • Microscopic haematuria (seen only on micro- Glomerular injury is caused by type II or type III scopic examination) with or without pro- antibody-mediated reactions, but the cause of teinuria

145 146 Chapter 12 • Autoimmune kidney disease Cryoglobulins Cryoglobulinaemic glomerulonephritis Membrano- proliferative C3 Nephritic factor Nephritic C3 glomerulonephritis 1 . disease Unknown Merck Manual Membranous Immune complex glomerulonephritis IgA complexes IgA immune nephropathy antibodies Antinuclear erythematosus Systemic lupus Autoimmune kidney disease syndrome Anti-GBM Goodpasture’s autoantibodies (ANCA) vasculitis cytoplasmic Antineutrophil autoantibodies Autoimmune kidney diseases and their immunological markers. Adapted from the ANCA associated Figure 12.1 Principles of treatment 147

• Macroscopic haematuria (visible with the fragile water-logged skin and impaired immune naked eye) function. • Nephrotic syndrome • Nephritic syndrome • Rapidly progressive glomerulonephritis Nephritic syndrome • Chronic glomerulonephritis. Nephritic syndrome develops as a consequence of glomerular inflammation. This leads to a Microscopic haematuria reduction in glomerular filtration rate (GFR), non-nephrotic proteinuria, haematuria, oedema Haematuria is caused by small breaks in the and hypertension. glomerular basement membrane that allow leak- age of red blood cells into the urine. Proteinuria may result from alterations in glomerular per- Rapidly progressive glomerulonephritis meability or tubulointerstitial damage. Micro- scopic haematuria (>10 106 red cells/L) and Rapidly progressive glomerulonephritis (RPGN) non-nephrotic proteinuria (<3 g/day) may occur is characterised by signs of glomerular inflam- in isolation or together but renal function mation and a rapid decline in renal function remains normal with no evidence of systemic over weeks or even days. Patients may present disease. When haematuria and proteinuria as an emergency requiring dialysis. Many occur together the risk of progressive renal dys- patients with RPGN have a systemic auto- function is much greater and warrants close immune disease. The histological hallmark is monitoring and possibly renal biopsy. glomerular crescents, giving rise to the name ‘crescentic glomerulonephritis’. Crescents are so-called due to their appearance on histological Macroscopic haematuria cross-section and are a poor prognostic sign. They develop from the proliferation of epithe- In episodic macroscopic haematuria urine is lial cells and mononuclear phagocytes in often a smoky brown colour rather than red Bowman’s capsule. and clots are unusual. Macroscopic haematuria is typically painless but may be accompanied by loin pain secondary to renal capsular Chronic glomerulonephritis swelling. Chronic glomerulonephritis is characterised by progressive renal insufficiency, glomerular Nephrotic syndrome inflammation, haematuria and frequently, hypertension. This term describes the chronic Nephrotic syndrome is characterised by severe kidney disease that ensues with long-standing proteinuria (>3 g/day) and oedema. Low serum glomerulonephritis, such as that in IgA albumin occurs secondary to protein loss, caus- nephropathy. ing a fall in plasma oncotic pressure and exacer- bating oedema. Low serum albumin also causes the liver to increase production of low-density Principles of treatment lipoproteins (LDL) with consequent hyperlipid- aemia. Platelet aggregation is enhanced, causing a hypercoagulable state which increases the risk The treatment of autoimmune kidney disease of venous thromboembolism. Kidney function consists of both supportive and disease specific may be normal, but may deteriorate if nephrotic therapies. Supportive therapies are those used to syndrome is prolonged. Nephrotic patients are treat most forms of kidney dysfunction and aim also at increased risk of infection partly due to to preserve renal function by reducing pro- 148 Chapter 12 • Autoimmune kidney disease teinuria, controlling oedema, and treating tion of granules containing cytolytic hypertension. Disease-specific therapies include enzymes to the surface of the neutrophils. various immunosuppressive therapies and 2 ANCAs then bind to the enzymes (antigens) plasma exchange in an attempt to suppress the on the surface of the neutrophils, most com- autoimmune disease itself. monly myeloperoxidase (MPO) or proteinase- 3 (PR3). The formation of this ANCA–enzyme complex causes activation of neutrophils which then bind to blood vessel walls. Antineutrophil cytoplasmic 3 Degranulation then occurs, causing release of autoantibody (ANCA)-associated cytolytic enzymes which in turn causes vasculitis inflammation and injury to the blood vessel wall. The resultant inflammation results in The vasculitides are a heterogeneous group of apoptosis of neutrophils and vessel endothe- uncommon diseases characterised by inflam- lial cells. matory cell infiltration and necrosis of blood ANCA is a sensitive marker and is raised in vessel walls. Vasculitis affecting the kidneys is 80–90% of patients with ANCA-associated vas- largely associated with autoantibodies against culitides but may also be raised in anti-GBM dis- components of the cytoplasm of neutrophils ease, inflammatory bowel disease, rheumatoid known as antineutrophil cytoplasmic auto- arthritis, hepatitis, bacterial endocarditis and antibodies (ANCA). ANCA-associated vascu- cystic fibrosis, though the titre is generally less litides include the syndromes Wegener’s marked. ANCA may rise and fall with disease granulomatosis, Churg–Strauss syndrome and activity but must be interpreted together with microscopic angitis, all of which can also affect clinical parameters and treatment should not be alveolar capillaries causing pulmonary haemor- initiated purely based on the ANCA titre. A rise rhage and dermal venules, causing purpura. in ANCA should, however, prompt clinical There are many other types of vasculitis not evaluation. Immunofluorescence assay produces associated with ANCA, though renal impair- two major staining patterns: a diffuse cyto- ment is usually less common and less severe. plasmic ANCA (c-ANCA) and the more concen- Vasculitis associated with IgA deposition trated perinuclear ANCA (p-ANCA). ANCAs are (Henoch–Schönlein purpura) and cryoglobuli- further tested by enzyme immunoassay to deter- naemia are discussed later under their respective mine their specificity for the enzymes PR3 and headings. Vasculitis may also occur with the MPO. Most c-ANCAs have specificity for PR3 following drug treatment: penicillamine, hydra- and most p-ANCAs for MPO, though this asso- lazine and propylthiouracil. Rarely, leukotriene ciation is not absolute. c-ANCA/PR3-ANCA is antagonists such as montelukast can cause a more commonly associated with Wegener’s Churg–Strauss-like syndrome, and this usually granulomatosis, p-ANCA/MPO-ANCA is more occurs on reduction or withdrawal of oral commonly associated with Churg–Strauss syn- steroid therapy. drome, and microscopic angitis is commonly associated with either.

Pathogenesis and antineutrophil cytoplasmic autoantibodies Epidemiology Antineutrophil cytoplasmic autoantibodies are ANCA-associated vasculitides most frequently thought to have a pathogenic role in the devel- present from mid-life onwards though may opment of necrotising vasculitis. The most com- occur at any age. They have a slight male pre- mon theory is detailed below: ponderance and affect people from any race. 1 A viral infection stimulates cytokine release The incidence is 1–2 per 100 000 population per which ‘primes’ neutrophils causing migra- year. Wegener’s granulomatosis may be more ANCA-associated vasculitis 149 frequent in colder climates, with the opposite tion of bone causing septal deformity requires being true for microscopic angitis.2 granuloma so does not occur in microscopic angitis. Mononeuritis multiplex is the most common Clinical manifestations neurological manifestation and is more frequent in Churg–Strauss syndrome. Gastrointestinal Renal involvement is common in ANCA-associ- involvement may present as abdominal pain, ated vasculitides and usually presents as rapidly blood in the stool, mesenteric ischaemia and progressive glomerulonephritis. Table 12.1 rarely perforation. The liver and pancreas may shows the relative organ systems involved in also be affected. the ANCA-associated vasculitides.2 Vasculitis may also present with generalised symptoms secondary to inflammation such as Diagnosis fever, malaise, anorexia, arthralgias and com- monly a flu-like syndrome. Skin involvement is All ANCA-associated vasculitides are capable of usually purpura secondary to dermal venulitis producing clinically indistinguishable symp- and is most commonly observed on the lower toms, so thorough clinical evaluation together extremities. Nodular lesions are common in with immunology screen and renal biopsy are Wegener’s granulomatosis and Churg–Strauss required. Wegener’s granulomatosis is char- syndrome but rare in microscopic angitis. acterised by granulomatous inflammation Nodules are caused by dermal or subcutaneous involving the respiratory tract and necrotising arteritis and by necrotising granulomatous glomerulonephritis is common. Churg–Strauss inflammation. syndrome is characterised by eosinophil-rich Upper and lower respiratory tract symptoms and granulomatous inflammation affecting the are more common in Wegener’s granulomatosis respiratory tract and is associated with asthma and Churg–Strauss syndrome. These conditions and eosinophilia. Microscopic angitis is char- can also have pulmonary haemorrhage second- acterised by necrotising glomerulonephritis ary to necrotising granulomatous inflammation. and pulmonary capillaritis but is distinguished By definition, microscopic angitis does not have from Wegener’s granulomatosis and Churg– granulomatous lesions. Upper respiratory mani- Strauss syndrome as granulomatous lesions are festations include sinusitis and rhinitis and are absent, as is eosinophilia. Glomerulonephritis is more common in Wegener’s granulomatosis. less common in patients with Churg–Strauss Ocular inflammation may also occur. Destruc- syndrome.

Table 12.1 The relative organ involvement in ANCA-associated vasculitides

Organ % Incidence

Microscopic Wegener’s Churg–Strauss angitis granulomatosis syndrome

Kidney 90 80 45 Skin 40 40 60 Lungs 50 90 70 Musculoskeletal 60 60 50 CNS 30 50 70 Gastrointestinal 50 50 50 ENT35 90 50

Adapted from ref. 2. 150 Chapter 12 • Autoimmune kidney disease

Prognosis of ANCA-associated vasculitides Induction Induction treatment is given for several months Without treatment, the majority of patients to patients with severe acute disease which would die within a year but with the advent of needs to be aggressively treated to prevent rapid immunosuppressive regimens 84% now survive progression of the disease. Treatment aims to a year and 76% survive for 5 years. End stage induce remission using relatively high doses of renal failure (ESRF) develops in up to a third of cyclophosphamide and prednisolone and usu- patients, but the most life-threatening compli- ally takes 3–6 months. Induction therapy may cation remains pulmonary haemorrhage. The be continued for a further three months post higher the serum creatinine at initiation of remission to prevent early relapse. Prednisolone treatment, the more likely it is that treatment dosages may be tapered during induction to will fail. Other poor prognostic signs include reduce adverse effects. Although this combina- age over 60 years and rapidly progressive tion is very effective, it is limited by long-term glomerulonephritis.3 toxic effects, requiring alternative approaches for maintenance therapy. Treatment of ANCA-associated vasculitides Corticosteroids Due to the high relapse rates in ANCA- In rapidly progressive disease pulsed IV methyl- associated vasculitides, long courses of immuno- prednisolone is given at a dose of 1 g daily for suppressive therapy are required which have 3 days then converted to oral prednisolone associated complications. This toxicity is an 1 mg/kg/day. Pulsed methylprednisolone may added burden on the already immuno- be omitted in less severe disease. Efficacy relates compromised patient with renal impairment. to the suppression of the acute and chronic Treatment consists of three phases: induction, inflammatory processes and immune cell func- maintenance and treatment of relapses. A sum- tion. Reversible short-term adverse effects mary of immunosuppressive regimens is shown include sodium and water retention, hyperten- in Table 12.2. sion, hyperglycaemia, CNS stimulation, peptic

Table 12.2 Immunosuppressive regimens in autoimmune kidney disease

Disease Induction therapy Maintenance therapy

ANCA-associated vasculitis IV cyclophosphamide 15 mg/kg every 2 Azathioprine 2 mg/kg/day weeks for three doses then monthly for six PLUS months low-dose prednisolone 5–10 mg/day PLUS IV methylprednisolone 1 g/day for 3 days then oral prednisolone 1 mg/kg/day Goodpasture’s syndrome Plasma exchange Continue oral cyclophosphamide PLUS PLUS oral cyclophosphamide 2 mg/kg/day low-dose prednisolone 5–10 mg/day PLUS IV methylprednisolone 1 g/day for 3 days then oral prednisolone 1 mg/kg/day Lupus nephritis IV cyclophosphamide 500 mg every 2 Azathioprine 2 mg/kg/day weeks for 3 months OR PLUS mycophenolate mofetil 1 g twice daily oral prednisolone 0.5–1 mg/kg/day ANCA-associated vasculitis 151 ulceration and immunosupression. Prolonged with IV cyclophosphamide therapy as it in- administration can lead to other adverse effects activates acrolein, thus preventing urothelial including osteoporosis, cataracts, skin fragility, toxicity. The dose of oral mesna is 40% of the myopathy, Cushingoid facies, hirsutism, alope- cyclophosphamide dose and is given at 2 hours cia and fat redistribution. The incidence of any before, 2 hours after and 6 hours after the adverse events increases with cumulative corti- cyclophosphamide infusion. costeroid dose. Cyclophosphamide can also lead to nausea Gastroprotection with a proton pump and vomiting, myelosuppression with neutro- inhibitor should be considered for all patients penia, infections due to immunosuppression, on corticosteroid therapy but particularly for alopecia and infertility. Prophylactic therapies those with additional risk factors such as should be used where appropriate as discussed advancing age and concomitant NSAID therapy. below. The incidence of leukaemia and lym- Osteoporosis prophylaxis should also be consid- phoma is increased with prolonged administra- ered using calcium and vitamin D supplements tion. Sperm banking should be offered to adult and bisphosphonates. Bisphosphonates have male patients, where appropriate, before any been shown to reduce both bone loss and frac- exposure to cyclophosphamide. Egg retrieval and ture rates in patients on corticosteroid therapy.4 preservation methods in female patients are still Although bisphosphonates are unlicensed in under investigation and are currently not routine patients with a creatinine clearance less than practice. One major drawback of egg retrieval is 30–35 mL/min, they are used in practice, with the time required to promote egg proliferation, weekly alendronate or risedronate being the which is often not compatible with the urgent agents of choice. HRT including tibolone may clinical need for immunosuppressive treatment. be considered in postmenopausal females. A suggested regimen is IV cyclophosphamide 15 mg/kg every two weeks for three doses then Cyclophosphamide monthly for six months. The cyclophos- Cyclophosphamide is a prodrug converted by phamide dose is generally reduced by about hepatic microsomal enzymes to the alkylating 25% in moderate renal impairment (creatinine agents 4-hydroxy-cyclophosphamide and phos- clearance 10–20 mL/min) and 50% in severe phoramide mustard, which alkylate guanidine renal impairment (<10 mL/min). Some units nucleotides thus blocking cell division. further reduce doses in elderly patients as Significant dose reductions are required in renal they are more likely to suffer neutropenia and impairment as 5–25% is excreted unchanged in other adverse events. A full blood count should the urine. be checked 10 days after the pulse is given Intravenous therapy is the treatment of as this is the nadir for bone marrow suppres- choice in most units although there is no con- sion. Subsequent doses should be reduced or sensus on a specific dosing regimen. A meta- withheld if the white cell count drops below analysis compared pulsed IV cyclophosphamide 4 109/L or platelet count drops below to daily oral cyclophosphamide and concluded 100 109/L. Concomitant allopurinol should that there was no difference in mortality or pro- be avoided as it can increase cyclophosphamide gression to ESRF. There was, however, a higher levels and the risk of myelosuppression. relapse rate following IV therapy and more adverse events following daily oral therapy.5 Methotrexate Oral treatment tends only to be used when IV The NORAM trial compared daily oral therapy is not possible or there has been good cyclophosphamide with weekly methotrexate response to IV therapy and oral therapy is more and adjunctive prednisolone in patients with convenient for the patient. Wegener’s granulomatosis and renal involve- Bladder toxicity is caused by renal excretion ment. There was no difference between groups of the metabolite acrolein which can cause in achieving remission, but patients with multi- haemorrhagic cystitis and an increased risk of organ involvement and those with pulmonary bladder cancer. Mesna is used prophylactically disease took longer to achieve remission with 152 Chapter 12 • Autoimmune kidney disease methotrexate.6 Overall relapse rate was also increased incidence of skin cancers and lym- higher in the methotrexate group. In practice, phomas following administration with pro- methotrexate is little used in patients with renal longed use. Patients being treated with impairment due to its high renal excretion azathioprine should limit their sun exposure (80–90%), which increases the risk of severe and use high-factor sun block when out in the toxicity. sun. Elimination of azathioprine requires hepatic metabolism by xanthine oxidase, Infliximab and rituximab leading to a significant interaction with the Infliximab, a monoclonal antibody that binds to xanthine oxidase inhibitor allopurinol. tumour necrosis factor alpha, and rituximab, a Concomitant therapy with allopurinol results in monoclonal antibody directed against the CD20 an increase in azathioprine levels of up to two- antigen on the surface of B-lymphocytes, have thirds, increasing the risk of toxicity unless the shown some promise as induction therapy in the azathioprine dose is reduced significantly. treatment of ANCA-associated vasculitides where Mycophenolate may be a useful alternative therapy with cyclophosphamide and steroids has when azathioprine is not tolerated or when failed. Evidence so far is limited to small studies concomitant allopurinol is required. and case reports for both agents. Further evid- ence is required to define the role of these new Plasma exchange agents in clinical practice and to determine their The MEPEX trial involved patients with safety profiles in this patient group. Wegener’s granulomatosis or microscopic angitis presenting with rapidly progressive glomerulonephritis and investigated whether Maintenance and relapse adjunctive plasma exchange was superior to The maintenance phase usually starts at least methylprednisolone pulses. All patients were 3–6 months after initial treatment at which treated with daily oral cyclophosphamide and point cyclophosphamide therapy is usually con- prednisolone and received maintenance therapy verted to azathioprine to reduce long-term toxic with azathioprine for a further six months. effects. Azathioprine therapy may be tapered Recovery of renal function was more frequent gradually and it may be possible to stop predni- with plasma exchange (67%) compared with the solone therapy altogether. If the patient’s con- methylprednisolone group (49%).6 The mech- dition relapses, treatment may be stepped back anism of plasma exchange may relate to the up and induction therapies added depending on removal of ANCA. It is still unknown whether the severity of the relapse. Additional therapies plasma exchange is helpful in other groups of such as plasma exchange or IV immuno- patients such as those with pulmonary haemor- globulins may also be required. rhage or less severe disease. In practice plasma exchange tends to be used in combination with Azathioprine and mycophenolate methylprednisolone. In the CYCAZAREM trial, patients received daily oral cyclophosphamide to induce remission, Immunoglobulins then one group continued cyclophosphamide Intravenous immunoglobulins may occasionally therapy while the other group was converted to be used as an additional agent for patients with azathioprine. There was no difference in relapse relapsing or refractory disease. Immuno- rate and adverse effects were comparable.7 modulatory doses of immunoglobulins used to Azathioprine is converted in the liver to treat autoimmune conditions are considerably 6-mercaptopurine, which inhibits purine larger than those used to treat immuno- biosynthesis. Azathioprine also suppresses deficiency states. The usual dose is inflammatory response and lymphocyte func- 400 mg/kg/day for 5 days. Some preparations tion. Adverse effects include nausea and vomit- contain sucrose, which can rarely induce ing, dose-dependent myelosuppression and osmotic injury to renal tubules and acute renal reversible cholestatic hepatotoxicity. There is an failure. Patients who may be at increased risk are Goodpasture’s syndrome 153 those with pre-existing renal impairment, dia- women in their 70s. Mortality without treat- betes, volume depletion, sepsis, parapro- ment is 90–95% but with aggressive treatment teinaemia, those over 65 years and those this falls to 10–20%. The disease occurs in any receiving concomitant nephrotoxic drugs. race but is most common in white Europeans.9

Adjunctive treatments Anti-infectives are often used prophylactically Clinical manifestations in patients receiving cyclophosphamide ther- apy, and although evidence is lacking, most units The diagnosis of Goodpasture’s syndrome is use prophylactic antibiotics to some extent. The dependent on clinical symptoms, the presence most commonly used is co-trimoxazole for pro- of anti-GBM antibodies and the absence of other phylaxis against Pneumocystis jiroveci (previously antibodies such as ANCA. It usually presents known as P. carinii) pneumonia (PCP). If co- with haemoptysis, dyspnoea and rapidly pro- trimoxazole is contraindicated then dapsone gressive renal failure. Pulmonary haemorrhage may be used and both of these require dose may precede renal failure by months or years adjustment dependent on renal function. Co- and almost exclusively occurs in smokers. trimoxazole may reduce upper respiratory Pulmonary haemorrhage is usually precipitated symptoms in Wegener’s granulomatosis but by infection and varies from mild haemoptysis does not affect relapse rate.8 Fungal prophylaxis to profound haemorrhage, leading to respiratory is used in some units and varies from nystatin failure. Haemoglobin should be monitored as a mouthwash to oral fluconazole. fall may indicate active bleeding. Pulmonary haemorrhage can also occur in systemic lupus erythematosus, rheumatoid arthritis, micro- Goodpasture’s syndrome scopic polyarteritis and Wegener’s granuloma- tosis, but these can usually be distinguished by Goodpasture’s syndrome is a rare hypersensitiv- specific antibody tests and renal biopsy. ity disorder of unknown cause, characterised by As with pulmonary haemorrhage, renal fail- circulating anti-GBM antibodies, pulmonary ure may also occur in isolation. Renal biopsy haemorrhage and rapidly progressive glomerulo- shows rapidly progressive glomerulonephritis nephritis. Goodpasture’s syndrome is caused by with crescent formation (crescentic glomerulo- antibodies against specific types of collagen in nephritis). Immunofluorescent staining shows the glomerular and alveolar basement mem- linear deposition of immunoglobulins and com- branes (anti-GBM antibodies). The antibody plement in the GBM and sometimes in the binds to these membranes, triggering destructive alveolar–capillary basement membrane. Patients inflammation which causes rapidly progressive presenting with severe renal injury with cres- glomerulonephritis, often with pulmonary haem- cents on biopsy may not respond to treatment. orrhage. Cigarette smoking and inhalation injury are thought to prime capillaries, increasing their susceptibility to damage by anti-GBM antibodies. Treatment

If Goodpasture’s syndrome is treated early, most Epidemiology patients recover and relapse is uncommon. If left untreated, however, most patients die Goodpasture’s syndrome is rare, with an estim- rapidly from pulmonary haemorrhage or renal ated incidence of 1 per 2 million in white failure. Assisted ventilation and haemodialysis European populations. It accounts for 5% of all are often required in the acute stage. Subsequent cases of glomerulonephritis and 10–20% of management requires pulsed methylpred- rapidly progressive glomerulonephritis cases. nisolone with oral cyclophosphamide and Patients with Goodpasture’s syndrome are repeated plasma exchange to remove circulating usually young men in their 30s or elderly anti-GBM antibodies. Platelet count should be 154 Chapter 12 • Autoimmune kidney disease monitored as plasma exchange can lead to con- and hormonal factors may be implicated in pro- sumption of platelets. Fresh frozen plasma ducing immunological defects. These defects should be administered at the end of plasma give rise to antibodies to nuclear components of exchange if the patient is at risk of bleeding. cells, termed antinuclear antibodies (ANAs). In Intensive daily plasma exchange is required to contrast to Goodpasture’s syndrome, ANAs are adequately reduce circulating antibodies and less specific and result in the deposition of restore renal function. Improvement is usually immune complexes throughout the body, evident within a few days of treatment. including the glomeruli, vasculature and skin Maintenance immunosuppressive therapy tissue. The deposition of immune complexes usually continues in the form of oral cyclo- causes an inflammatory response in the affected phosphamide 2 mg/kg/day and prednisolone tissue. 1 mg/kg/day (Table 12.2). Prednisolone therapy is maintained for at least four weeks then slowly tapered to 5–10 mg daily, which should be Diagnosis continued until treatment is withdrawn. Oral cyclophosphamide may be tapered by 25 mg The fluorescent test for ANA is positive and high every 2–3 months according to disease activity. in titre in the vast majority of SLE patients, Duration of treatment varies considerably and though the presence of ANA is not limited to may be required for up to 18 months in some SLE. Anti-double-stranded DNA antibodies patients. Some patients progress to ESRF and (anti-dsDNA) are a specific diagnostic marker for require dialysis or transplantation. Recurrence SLE and are present in most patients. Disease of Goodpasture’s syndrome after kidney trans- activity may be affected by hormonal mechan- 9 plantation is rare. isms, with flares very rare after the menopause and hormone replacement therapy (HRT) increasing flares in some patients. Drug-induced Systemic lupus erythematosus causes of SLE flares such as hydralazine should be borne in mind. About a third of patients with lupus have Systemic lupus erythematosus (SLE) is a chronic antiphospholipid antibodies (APAs) in their cir- inflammatory connective tissue disorder of culation, which may increase the coagulability unknown cause that can affect almost any of blood. This increases the risk of arterial and organ or system in the body. When SLE affects venous thrombosis, low platelet count, miscar- the kidneys it is termed lupus nephritis. SLE fol- riages and a skin rash called livedo reticularis. lows a relapsing–remitting course which varies widely from rapidly progressive to relatively benign, often with long periods of remission, Clinical manifestations interspersed with relapses termed ‘flares’. SLE affects at least 12 in 100 000 people in the UK, Systemic lupus erythematosus may present and 90% of cases occur in women. It usually abruptly with fever or develop insidiously over presents between the ages of 15 and 40 and is months or years with episodes of fever and more prevalent in Afro-Caribbean and Asian malaise. It is a multisystem disease which can populations than in the white population. affect almost any part of the body. The American Prognosis is usually good provided acute College of Rheumatology suggests that a diag- episodes are treated aggressively.11 nosis of SLE is likely when four or more of the following clinical features are present:2 Pathogenesis • Malar rash (‘butterfly’ rash across the cheeks) • Discoid rash The cause and pathogenesis of SLE is not com- • Skin photosensitivity pletely understood but genetic, environmental • Oral ulcers Systemic lupus erythematosus 155

• Non-erosive arthritis Lupus nephritis flares may present as • Serositis nephrotic or nephritic syndrome. Nephritic syn- • Renal involvement (proteinuria or haema- drome is often accompanied by low comple- turia) ment levels and rising anti-dsDNA antibodies. • Neurological disorder (seizures, psychosis) Lupus nephritis flares are common despite • Haematological disease (leucopenia, throm- treatment and occur in up to two-thirds of bocytopenia, lymphopenia) patients. They are more common if steroids are • Immunological disorder (anti-dsDNA anti- used alone compared with when immuno- bodies, APAs) supression with cyclophosphamide or myco- • Raised antinuclear antibodies (ANAs). phenolate is used. Other risk factors for lupus nephritis flares include young age, male gender, black race, severe SLE, delay in initiating treat- Lupus nephritis ment, delay in achieving remission, partial response to treatment and rising anti-dsDNA Around 50% of patients with SLE develop clinic- antibodies. Renal function can be preserved if ally evident renal disease, though this is largely additional immunosuppressive therapy is benign with only a small minority of patients started early on relapse, though some patients developing ESRF.11 The World Health Organ- will progress to ESRF requiring dialysis or trans- ization has classified lupus nephritis based on plantation. the nature of glomerular lesion (Table 12.3 2). Glomerular lesions may change over time and usually more than one type of lesion is present. Treatment Classes I and II lupus nephritis have an ex- cellent prognosis and are monitored but not Treatment varies depending on the severity of specifically treated. Classes III and IV or pro- disease. Mild lupus nephritis associated with liferative lupus nephritis are more fulminant, classes I, III and V usually only requires symp- requiring aggressive treatment to induce remis- tomatic therapy and occasionally moderate sion and prevent significant renal morbidity doses of corticosteroids, for example 10–15 mg and mortality. Class IV has the worst prognosis of prednisolone. More severe lupus nephritis without treatment though most patients survive associated with class III or IV nephritis usually with aggressive treatment. Class V, or mem- requires immunosuppressive therapy. The goal branous lupus, is characterised by nephrotic of long-term immunosuppressive therapy is to syndrome but treatment regimens are less well suppress lupus with minimum side-effects. It is defined than classes III and IV. usually possible to stop treatment after several years provided the patient has stable renal func- tion, normal immunology and no proteinuria.

Table 12.3 WHO classification of lupus nephritis Treatment of extrarenal manifestations Category Histological picture Incidence The most common symptoms of SLE are fatigue Class I Normal glomeruli <10% and pain in the joints and muscles which can Class II Mesangial glomerulonephritis 10–20% be managed with NSAIDs and paracetamol. The Class III Focal proliferative 15–20% risks and benefits of NSAIDs in patients with glomerulonephritis renal impairment should be carefully assessed Class IV Diffuse proliferative 50% and if considered essential then renal function glomerulonephritis should be closely monitored. Gastroprotection Class V Membranous 15% should be considered, especially if the patient is glomerulonephritis taking NSAIDs regularly or with concomitant steroids. Hydroxychloroquine is often used for From ref. 2. cutaneous SLE or arthralgias not controlled by 156 Chapter 12 • Autoimmune kidney disease

NSAIDs and paracetamol. The dose is 200– units use IV pulsed cyclophosphamide of vary- 400 mg daily which may then be reduced once ing frequency and dose. The Euro-Lupus control is achieved. Hydroxychloroquine is Nephritis Trial compared a high-dose to a low- relatively safe and well tolerated. Though retinal dose cyclophosphamide regimen and found toxicity is rare, affecting 1 in 1800 patients, comparable results in treatment failure, renal patients should have annual eye tests and be remission, renal flares and side-effect profiles. referred to an ophthalmologist if there is any All patients received three pulses of methyl- change in visual acuity.12 Patients with high prednisolone followed by oral prednisolone levels of APAs should be prescribed aspirin 75– therapy. The low-dose cyclophosphamide proto- 150 mg daily as thromboprophylaxis. Patients col was 500 mg every two weeks for six doses with cerebral lupus and patients with APA who compared with the high-dose regimen of have already suffered thromboses or miscar- 500 mg/m2 monthly for six months then quar- riages should be considered for warfarin therapy terly for two pulses. Azathioprine 2 mg/kg/day as they are at risk of further thromboses. was started in all patients two weeks after the last cyclophosphamide pulse.13 Immunosuppression Azathioprine and mycophenolate Immunosuppressive therapy for SLE is depend- ent on disease severity. In lupus nephritis, treat- Azathioprine is usually used to replace ment is similar to that for systemic vasculitis but cyclophosphamide as maintenance therapy as regimens differ slightly based on evidence (see in the protocol above but it may also be used to Table 12.2). As in vasculitis, studies involving allow reduction of steroid dose in patients who infliximab and rituximab are limited, with ther- have recurrent flares when the prednisolone apy being reserved for patients with disease dose is reduced. Mycophenolate mofetil may be resistant to standard therapies. Concerns have an alternative to azathioprine and has been been raised that infliximab may not be suitable used in recent studies to induce remission. in SLE due to an increased risk of developing Mycophenolate acts by inhibiting lymphocyte ANA and worsening disease activity. proliferation, antibody formation and genera- tion of cytotoxic T-cells. Several trials invest- igating its use in lupus nephritis have shown Corticosteroids that mycophenolate may be at least as effective Mild SLE flares can be controlled with ‘one-off’ as oral cyclophosphamide and possibly more doses of intramuscular methylprednisolone effective than IV pulsed cyclophosphamide which may avoid long-term steroid use. More therapy.14 Although mycophenolate may cause acute flares may require a course of daily oral more nausea and diarrhoea, this is usually self- prednisolone starting at 20–30 mg daily, reduc- limiting and is appealing, considering the more ing slowly dependent on disease activity. Higher serious adverse effects with cyclophosphamide. doses of steroids are reserved for more severe Limitations of the above studies include vari- disease such as lupus nephritis and cerebral ations in type and extent of renal injury lupus, which require IV pulses of methyl- amongst patients and the limited follow-up prednisolone followed by oral prednisolone time, so further studies are warranted to confirm 1 mg/kg/day. Hirsutism and weight gain may be these findings. particularly troublesome given that most patients are younger females. IgA nephropathy Cyclophosphamide Cyclophosphamide is used in combination with IgA nephropathy is a form of glomerulo- steroids to treat the most severe forms of SLE nephritis characterised by deposition of including lupus nephritis. Most protocols in UK immunoglobulin A (IgA) in the GBM. IgA IgA nephropathy 157 nephropathy is highly variable and its clinical 15% of patients by 10 years and 20% by 20 course may vary from slow progression to rap- years.15 IgA nephropathy may be picked up on idly progressive glomerulonephritis. Although routine blood or urine analysis as raised plasma IgA nephropathy is a non-systemic disease only creatinine or proteinuria but usually presents affecting the kidney, IgA deposition may also be with either episodic macroscopic haematuria or associated with systemic illnesses, most com- persistent microscopic haematuria. monly Henoch–Schönlein purpura and SLE. Acute renal failure with oedema, hyperten- Henoch–Schönlein purpura is a small-vessel vas- sion and oliguria occurs in less than 5% of culitis affecting the skin, joints, gut and kidney. patients. It can present as rapidly progressive It has the same renal features of IgA nephro- glomerulonephritis or mild glomerular injury pathy but is differentiated by extrarenal features with macroscopic haematuria secondary to caused by tissue deposition of IgA. tubular damage. Chronic renal failure is usually slowly progressive, taking many years to develop to ESRF. Pathophysiology In IgA nephropathy, macroscopic haematuria is largely associated with upper respiratory tract Kidney biopsy shows deposition of IgA and infections and usually presents within 48–72 complement in the GBM indicating that this is hours after the infection begins. Macroscopic the result of deposition of immune complexes haematuria may also follow tonsillectomy, leading to the activation of the complement vaccination, strenuous physical exercise and cascade. On biopsy, glomeruli may be normal, trauma. Between episodes of gross haematuria but with long standing disease there are vary- many patients have persistent microhaematuria, ing degrees of glomerular sclerosis and tubulo- proteinuria or both. Microscopic haematuria is interstitial scarring which represent chronic usually associated with proteinuria and occa- kidney damage. Acute renal failure shows as sionally nephrotic syndrome. Patients with necrotising glomerulonephritis with crescent microscopic haematuria have a higher risk of formation and renal tubular occlusion with red progressive renal failure than those with macro- cell casts. scopic haematuria, possibly because they are identified later. Sustained hypertension, impaired renal function and proteinuria above Epidemiology 1 g/day are poor prognostic signs. The symp- toms and course of IgA nephropathy is Distribution of IgA nephropathy varies with influenced by the susceptibility of the glomeruli geographic area throughout the world and is to injury which is likely to have a genetic more common in Asia than in Europe and the component. USA. IgA nephropathy is more common in Henoch–Schönlein purpura usually begins white and Asian populations and is rare in Afro- with a palpable purpuric skin rash involving the Caribbean populations. It has a male predomin- feet, legs and arms. Most patients have a fever ance of at least 2:1 and can affect all ages, but accompanied with polyarthralgia with associ- is most common in the second and third decade ated tenderness and swelling of the ankles, of life.15 Henoch–Schönlein purpura is most knees, hips, wrists and elbows. Abdominal pain common in the first decade of life, though it and melaena may also be present secondary to may occur at any age. gut vasculitis. Up to half of patients will develop haematuria and proteinuria. Henoch–Schönlein purpura usually remits within four weeks of Clinical features onset, but frequently recurs after a disease-free period of several weeks. In most patients the dis- IgA nephropathy follows a benign course in order subsides without serious complications; most patients, but many still progress to end however, some patients go on to develop stage renal disease, which develops in about chronic renal failure. 158 Chapter 12 • Autoimmune kidney disease

Supportive treatment azathioprine. Renal transplantation is effective but IgA nephropathy recurs in 20–60% of Supportive treatment aims to prevent worsening patients and usually progresses slowly, with of renal function and concentrates on aggressive graft loss occurring in less than 10% of management of hypertension and proteinuria. patients.15 ACE inhibitors are the agent of choice to treat hypertension as they also have beneficial effects on proteinuria. Proteinuria should also be Treatment of Henoch–Schönlein purpura treated with ACE inhibitors in normotensive patients if tolerated as, in addition to their effect Treatment of Henoch–Schönlein purpura is on blood pressure, they may decrease protein- somewhat different, requiring only sympto- uria by decreasing intraglomerular pressure. matic management. Prednisolone may be used ACE inhibitors have been shown to preserve to treat arthralgias, abdominal pain and oedema renal function in patients with proteinuric IgA but has no effect on the course of renal disease. nephropathy.16 Angiotensin II receptor blockers Immunosuppressive regimens with cyclophos- can be used for patients who cannot tolerate phamide with prednisolone may be beneficial ACE inhibitors due to persistent cough. They in severe nephritis. Plasma exchange may also can also be added to maximal ACE inhibitor be beneficial in severe nephritis but there is therapy (unlicensed), which may have an addi- little evidence to confirm this. tive effect on reducing proteinuria, but whether this has further beneficial effects on renal func- tion is not yet known. Membranous glomerulonephritis

Immunosupressive treatment Membranous glomerulonephritis is a condition that mainly affects adults and is characterised The risk benefit for immunosuppressive regi- by the insidious onset of nephrotic syndrome. mens is often unfavourable due to the slowly Patients usually have normal renal function and progressive nature of IgA nephropathy, but may normal or elevated blood pressure. Membranous be beneficial in the small minority of patients glomerulonephritis is usually idiopathic but with rapidly progressive disease. Prednisolone may be secondary to other diseases such as SLE, may reduce proteinuria and preserve renal func- hepatitis, malaria and malignancies. Drugs may tion in those patients with preserved renal also precipitate membranous glomeruloneph- function and minimal glomerular injury, ritis, including gold, penicillamine, captopril though the benefit of prednisolone over aggress- and lithium. Membranous glomerulonephritis ive supportive therapy has not yet been is thought to be due to the deposition of proven.17 The immunosuppressive regimen of immune complexes in the GBM, though anti- choice is usually prednisolone monotherapy, as gens associated with idiopathic disease have not no clear benefit has been shown with other been identified. agents. Daily oral cyclophosphamide in addi- About a third of patients remit spontane- tion to prednisolone should be reserved for ously without further relapses. Another third those with rapidly progressive disease.18 Even develop chronic renal impairment and a further with immunosuppression, up to half of these third progress to ESRF. Patients usually present patient will progress to ESRF within 12 months. with oedema, proteinuria (>3 g/day) and occa- Patients may be switched to azathioprine once sionally haematuria and hypertension. Hyper- in remission, as in vasculitis and SLE, though tension may be secondary to deteriorating renal caution should be exercised as there have been function. Patients may also present with non- rare reports of anaemia and leucopenia with specific symptoms such as anorexia, malaise concomitant ACE inhibitor therapy and and fatigue. Cryoglobulinaemic glomerulonephritis 159

Supportive treatment Although it can be idiopathic, it more often occurs secondary to other diseases such as SLE, Asymptomatic patients should be monitored for cryoglobulinaemia, hepatitis and some malig- deterioration in renal function but are not nancies. Membranoproliferative glomerulo- actively treated as their long-term prognosis is nephritis is characterised by chronic immune good. Patients with oedema are treated sympto- complex deposition in the glomerulus causing matically with diuretics. Supportive therapies glomerulosclerosis. The antibodies thought to such as ACE inhibitors should be prescribed be involved in its pathogenesis are known as initially, adding in an angiotensin II receptor nephritic factors, most commonly C3 nephritic blocker if required, as in IgA nephropathy. factor.

Immunosuppressive treatment Clinical manifestations

Immunosuppressive therapies are reserved for Membranoproliferative glomerulonephritis pre- patients with severe symptomatic disease who sents with nephrotic syndrome in the vast are at most risk from disease progression. This majority of patients and is usually accompanied includes patients with severe nephrotic syn- by microscopic haematuria. Occasionally drome (proteinuria >10 g/day) and renal impair- macroscopic haematuria, acute renal failure and ment which cannot be controlled with hypertension occur. Kidney biopsy is required to supportive therapies. confirm diagnosis and differentiate the disease Prednisolone monotherapy is not generally from membranous nephropathy. In general the used as it has no effect on remission or preser- long-term prognosis is poor, with ESRF occur- vation of renal function. Monotherapy with ring in 50% of patients by 3–5 years and 75% daily oral cyclophosphamide has been shown to by 10 years. reduce proteinuria and preserve renal function and is given at a dose of 1.5–2 mg/kg/day. Chlorambucil is an alkylating agent which can Treatment be used in an alternative regimen as below: • Days 1–3: intravenous methylprednisolone Treatment strategies are variable due to the 1 g/day scarcity of randomised controlled trials. Support- • Days 1–28: chlorambucil (0.2 mg/kg/day) ive treatment is the same as that for membran- • Days 29–56: prednisolone (0.5 mg/kg/day). ous nephropathy, though there is little evidence to support prednisolone or other immunosup- This cycle is repeated for three cycles (six pressive therapy in these patients. Antiplatelet months in total). therapy is the only intervention shown to be of For patients intolerant of cytotoxic therapy, benefit in adult patients with membranopro- ciclosporin may be used at 4–6 mg/kg/day but liferative glomerulonephritis. The probable patients frequently relapse following with- mechanism for antiplatelet therapy is platelet drawal. There is currently no consensus on a inhibition and altered renal haemodynamics. 19 specific immunosuppressive regimen. Combination therapy with aspirin and dipyrid- amole is usually the drug regimen of choice.20

Membranoproliferative glomerulonephritis Cryoglobulinaemic glomerulonephritis Membranoproliferative glomerulonephritis is an uncommon cause of chronic glomeruloneph- Cryoglobulins are immunoglobulins that ritis mainly affecting children and young adults. undergo reversible precipitation when plasma is 160 Chapter 12 • Autoimmune kidney disease cooled while flowing through small blood ves- NSAIDs, though these must be used with cau- sels. Cryoglobulin immune complexes may tion in renal impairment. Patients with mild to deposit in any part of the body, resulting in moderate arthritis may respond to low doses of inflammation and vasculitis. Cryoglobulin- steroids with or without hydroxychloroquine. If aemic glomerulonephritis occurs when cryo- there is evidence of renal disease or vasculitis globulin immune complexes deposit in the immunosuppressive therapy may be required. glomerular capillaries. The survival rate for Combination therapy with oral cyclophos- patients with renal impairment is around 60% phamide and prednisolone are the agents of at 5 years from diagnosis. The occurrence of choice as discussed under ANCA-associated cryoglobulins may be idiopathic but is strongly vasculitis. Life-threatening disease requires associated with hepatitis C infection, and may plasma exchange in addition to immuno- also be seen in other autoimmune diseases suppression. including SLE. If there is a secondary cause then the main focus of therapy is to treat the underlying dis- ease. SLE should be treated as discussed earlier. Clinical manifestations Cryoglobulinaemic glomerulonephritis is associ- ated with hepatitis C infection in the vast Skin involvement is very common, including majority of patients and should be treated with purpura and Raynaud’s phenomenon, and is pegylated interferon and ribavirin, though due to the precipitation of immune complexes expert opinion should be sought.21 in the small blood vessels of the skin. Ischaemic necrosis of the skin may also occur. Arthralgia and myalgia are common but progressive arth- ritis is very rare. Pulmonary involvement may Conclusions result in dyspnoea and cough. Abdominal pain may be present secondary to vasculitis affecting Autoimmune kidney disease covers a wide range the gut. Arterial thrombosis, though rare, can of diseases which cause varying degrees of renal occur secondary to increased serum viscosity. dysfunction from mild renal impairment to Renal failure may present with isolated haema- life-threatening rapidly progressive glomerulo- turia, proteinuria or nephrotic syndrome and is nephritis. Rapidly progressive disease always a serious complication, following a progressive requires immunosuppressive therapies which course similar to that of membranoproliferative carry their own risks and managing the toxic glomerulonephritis. effects of these agents may be just as challeng- ing as managing the disease itself. Less severe disease may not require immunosuppressants Treatment at all, so the risks and benefits of treatment should be weighed carefully against the severity Patients with myalgia or arthralgia may be of disease. treated with paracetamol or, if necessary, References 161

C CASE STUDIES

Case 1 Mrs X is 74 years old and is admitted to the nephrology ward with acute renal failure (GFR <10 mL/min) and haemoptysis. A full immunology screen is positive for c-ANCA in high titre and the chest X-ray shows evidence of granulomatous lesions.

Q1. What diagnosis is most likely to be made?

Q2. What initial treatment would you recommend?

Q3. How would you monitor this therapy?

Q4. What adjunctive therapies would you consider for this patient?

Q5. What advice would you give to the patient regarding their new treatment regimen?

Case 2 Mr Y, aged 22 years, is admitted to the nephrology ward for control of hypertension. Urinalysis is positive for microscopic haematuria and proteinuria. On routine blood chemistry serum creati- nine is raised at 167 µmol/L.

Q1. What drug therapy would you suggest?

Q2. What is the target blood pressure for this patient? Six months later Mr Y is readmitted with persistent hypertension despite supportive therapy. A 24- hour urine collection is performed showing a protein excretion of 2 g/day and a creatinine clear- ance of 36 mL/min. The consultant advises a kidney biopsy which shows IgA and complement deposition in the GBM.

Q3. What is the prognosis for Mr Y?

Q4. What further drug therapy would you recommend?

References 4. Royal College of Physicians. Glucocorticoid-induced Osteoporosis. London: Royal College of Physi- cians, December 2002. 1. The Merck Manual, 17th edn. 1999, Section 17, 5. de Groot K, Dwomoa A, Savage C. The value of Chapter 224, p. 1857. pulse cyclophosphamide in ANCA associated 2. Feehally J, Johnson K. Comprehensive Clinical vasculitis: meta analysis and critical review. Nephrology, 2nd edn. London: Harcourt, 2003, Nephrol Dial Transplant 2001; 16: 2018–2027. Section 5 Chapters 26 and 27. 6. de Groot K, Jayne D. What is new in the therapy 3. Booth A, Almond MK, Burns A et al. Outcome of of ANCA-associated vasculitides? Clin Nephrol ANCA-associated renal vasculitis: a 5-year retro- 2005; 64: 480–484. spective study. Am J Kidney Dis 2003; 41: 7. Jayne D, Rasmussen N, Andrassy K et al. A 776–784. randomized trial of maintenance therapy for 162 Chapter 12 • Autoimmune kidney disease

vasculitis associated with antineutrophil cyto- 14. Ginzler E, Aranow C. Mycophenolate mofetil in plasmic autoantibodies. N Engl J Med 2003; 349: lupus nephritis. Lupus 2005; 14: 59–64. 36–44. 15. Brake M, Somers D. IgA Nephropathy. eMedicine 8. Stegeman C, Cohen Tervaert JW, de Jong PE, 2006, updated 23 August. Kallenberg CGM and the Dutch Co-trimoxazole 16. Praga M, Gutiérrez E, González E et al. Treatment Wegener Study Group. Co-trimoxazole for the of IgA nephropathy with ACE inhibitors: a ran- prevention of relapses of Wegener’s granulo- domized controlled trial. J Am Soc Nephrol 2003; matosis. N Engl J Med 1996; 335: 16–20. 14: 1578–1583. 9. Kluth D, Rees A. Anti-glomerular basement mem- 17. Pozzi C, Andrulli S, Del Vecchio L et al. brane disease. J Am Soc Nephrol 1999; 10: Corticosteroid effectiveness in IgA nephropathy: 2446–2453. long-term results of a randomized controlled 10. The Merck Manual of Diagnosis and Therapy, 18th trial. J Am Soc Nephrol 2004; 15: 157–163. edn. 2007, Chapter 5 Section 50. 18. Samuels J. Immunosuppressive agents for treating 11. Hildebrand J. Systemic lupus erythematosus. IgA nephropathy (review). The Cochrane Library, E-Medicine 2005; updated 21 November. 2006 Issue 3. 12. Rahman A. Drug treatment of systemic lupus 19. Ponticelli C, Passerini P. Treatment of membran- erythematosus. Hosp Pharm 2001; 8: 69–73. ous nephropathy. Nephrol Dial Transplant 2001; 13. Houssiau FA, Vasconcelos C, D’Cruz D et al. 16 (Suppl 5): 8–10. Immunosuppressive therapy in lupus nephritis: 20. Kathuria P. Membranoproliferative glomerulo- the Euro-Lupus Nephritis Trial, a randomized nephritis. eMedicine 2006; updated 11 April. control trial of low-dose versus high dose intra- 21. Edgerton C. Cryoglobulinaemia. eMedicine 2006; venous cyclophosphamide. Arthritis Rheum 2002; updated 26 January. 46: 2121–2131. 13

Paediatric kidney disease

Susan Patey

This chapter provides a guide to the particular most children with renal failure have been born challenges that face the paediatric renal phar- with abnormal kidneys (e.g. polycystic or dys- macist from the initial calculation of glomerular plastic kidneys). Less commonly, an illness can filtration rate to dosing and administration of cause damage to the kidneys during childhood medicines. It will deal with chronic renal failure (e.g. lupus nephritis, haemolytic uraemic syn- and its progression to end stage, dialysis and drome, Henoch–Schönlein purpura nephro- transplantation. pathy). Occasionally, there may be a genetic Many medicines are not approved for paedi- cause such as cystinosis, a metabolic disease or atrics either because explicit approval has not congenital nephrotic syndrome, which often been obtained by the manufacturers as studies requires bilateral nephrectomy. have not been performed in children or because As in adults, the markers for renal function are of toxic effects on development as observed in creatinine and glomerular filtration rate (GFR). animal experiments (e.g. quinolones). Some of these medicines, however, are used in vital indi- cations by paediatricians, thus licensed prepara- Creatinine and glomerular filtration tions may be used ‘off label’ and some rate preparations suitable for paediatrics may be made by specials manufacturers (unlicensed). The issue of licensing of medicines for chil- Creatinine is a very good marker of kidney func- dren is being addressed by the European tion, but because it comes from muscle, the Commission. In its section on medicines, the blood level is dependent on the size of the child National Service Framework for Children pub- in addition to how well the kidneys are work- lished in October 2004,1 envisages that all chil- ing. Therefore, the creatinine level in an adult dren and young people will receive medicines would be higher compared with that in a child that are safe and effective in formulations that and, similarly, a higher level would be found in can be easily administered, are appropriate to a child than in a baby. One would expect the their age and have minimum impact on their creatinine to rise throughout childhood until education and lifestyle. Children, young people adulthood is reached. For this reason the and their parents or carers need to be well- Cockroft and Gault equation for calculating informed and supported to make choices about GFR is unsuitable for use in paediatric patients. their medicines and to become competent in An alternative formula which provides a good the administration of medicines. Children with approximation of GFR in children is that chronic renal failure are often required to take a derived by Morris et al. in 1982 using large number of different medicines, which are height/serum creatinine:2 then replaced by another set of medicines fol- lowing a renal transplant. GFR (mL/min) 40 height (cm) The causes of chronic kidney disease in chil- (corrected to = Serum creatinine (µmol/L) dren are very different to those in adults, as 1.73 m2)

163 164 Chapter 13 • Paediatric kidney disease

(Creatinine level of 1 mg/mL = 88.5 µmol/L) many years, or even improve, especially in the This equation cannot be applied to infants and first year of life. Abnormal kidneys may not be neonates. able to grow as the child grows, and therefore As the Morris equation is corrected for body become progressively smaller relative to the surface area, the resultant GFR in mL/min does child’s body size. This may be particularly the not require further manipulation when making case during puberty, when the child is growing dosage decisions based on references that utilise very fast, so this may be a time when kidney GFRs that have been normalised to 1.73 m2 (e.g. function declines more rapidly. Many children Guy’s and St Thomas’ Paediatric Formulary, with abnormal kidneys are unable to concen- British National Formulary for Children, trate their urine so pass very large volumes. Daschner’s paediatric dosing tables).3 They therefore need to drink a lot to compen- Others have identified methods of estimating sate, particularly if they have extra fluid losses GFR in children without the use of height or such as with diarrhoea, vomiting or fever. weight.4 The normal range of GFR is 80–120 mL/ 2 min/1.73 m . A GFR between 60 to 80 is usually Monitoring and treatment of children considered to indicate mild chronic renal failure with chronic renal failure (CRF), 30–60 moderate, and below 30 severe CRF. When the GFR falls below 15 mL/min/ 1.73 m2, dialysis or transplantation is usually Nutrition and growth necessary. GFR is low at birth, averaging 3 mL/min/ Chronic renal failure can affect growth, mainly 1.73 m2 in the term baby, rapidly increases dur- due to poor nutrition, which may be because of ing the first six months of life and then reduced appetite or vomiting. All infants and increases gradually to adult values by 1–2 years. children CRF should be referred to a dietician. In the neonatal period the GFR may be stan- The aim is produce a diet that will control dardised to body weight as this correction pro- symptoms and prevent complications, particu- duces less variability (Table 13.1). larly uraemia and renal bone disease, and will Usually when both kidneys are affected and promote optimum growth but will preserve the GFR is below normal, the kidney function residual renal function. Tube feeding or a gas- gets progressively worse, although this does not trostomy is often required to supplement oral always happen and it may remain stable for intake. Height, weight and head circumference

Table 13.1 Approximate GFR based on age, weight and body surface area

Weight (kg) BSA (m2) GFR

mL/min mL/min/1.73 m2

Premature infants 1.0 0.1 0.2 3.5 Newborns 2.0 0.15 0.5 6.0 3.2 0.2 1.5 13.0 1 month 4.0 0.25 7.5 52.0 3 months 6.0 0.35 11.0 54.0 1 year 9.2 0.45 30.0 115.0 10 years 30.0 1.0 70.0 120.0 Adults 70.0 1.73 120.0 120.0

BSA, body surface area; GFR, glomerular filtration rate. From ref. 5. Monitoring and treatment of children 165 must be plotted on a growth chart to monitor of any proteinuria, will determine which group growth and development. If vomiting is a prob- is used initially. If the GFR is <10 mL/min/ lem, domperidone may be prescribed at a dose 1.73 m2 always start with the lowest dose. of 200–400 µg/kg three to four times a day, For infants (less than 1 year old) use shorter although this should be used with caution in acting agents for flexibility of dosage: propran- infants under one month of age due to the pos- olol instead of atenolol; captopril instead of sibility of extrapyramidal side-effects (this indi- enalapril (Table 13.3). Once stable, the patient cation is unlicensed for use in children). Very may be changed to the longer acting anti- rarely, if growth is poor despite optimum nutri- hypertensive agent. tion, growth hormone may be prescribed.

Proteinuria Blood pressure The presence of protein in the urine has been Blood pressure varies with age. It can be low in shown to increase the rate of decline of renal some children with CRF as their kidneys ‘leak’ function. An ACE inhibitor or angiotensin II salt and water. These children usually need to receptor blocker may be prescribed. drink a lot and may need sodium chloride sup- plements. A 1 mmol in 1 mL solution is avail- Bicarbonate able as a ‘special’ and should be given orally, initially at a dose of 1–2 mmol sodium/kg/day The kidneys are responsible for regulating the in 3–4 divided doses and adjusted as necessary. acidity of the blood. Sodium bicarbonate may Children who are salt losers or able to deal need to be prescribed. For chronic replacement with salt normally may develop problems as a 1 mmol in 1 mL solution is available as a ‘spe- their renal function declines. cial’. The initial oral dose is 1–2 mmol/kg/day Some children are unable to lose salt and for infants and 70 mmol/m2/day for older chil- water as they should and consequently will dren, given in 2–4 divided doses and adjusted need to reduce their salt intake. These children according to plasma levels. are likely to develop high blood pressure and will need appropriate treatment (diuretic, cal- cium channel blocker, angiotensin-converting Urea enzyme (ACE) inhibitor etc.). The measurement of blood pressure in chil- Urea is produced when protein in the diet is dren, particularly the younger age group, broken down so that it can be used in the body requires practice and specialist paediatric skills for growth. The urea level will therefore depend and knowledge. The US Task Force report on upon the protein and calorie intake in the diet. blood pressure is a good reference source, which It will rise if the protein intake is very high or takes into consideration the child’s gender, age the calorie intake is very low, as this reflects the 6 and height percentiles. breakdown of protein from muscle as a source The focus here is to use medicines that of energy. A high urea level can make a child preferably can be used once daily, maximising feel very ill. The dietitian will provide advice treatment dosage before adding a further medi- on controlling the urea level. The aim is to cine. Compliance in infants and children can be keep plasma levels below 20 mmol/L or below a particular problem as they are often on multi- 30 mmol/L in children over 10 years of age. ple medicines. The agent(s) used will be from the following groups: ACE inhibitor; beta- blocker; calcium channel blocker; diuretic. A Calcium, phosphate and parathyroid hormone beta-blocker should not be used alone as first- line therapy (Table 13.2). The kidneys are responsible for maintaining a The clinical situation, including the presence healthy balance of calcium, phosphate and vita- 166 Chapter 13 • Paediatric kidney disease

Table 13.2 Long-term treatment of hypertension in children and adolescents as used at Great Ormond Street Hospital, London

Drug Route Normal Normal and Divided Preparations starting maximum doses in and comments dose dose range a day

Amlodipine Oral 100–200 µg/ 1.25 mg (body weight 1 Tablets 5 mg, 10 mg kg per dose 6–15 kg) Tablets may be dispersed in 2.5 mg (body weight water and still maintain long 15–25 kg) acting effect 5 mg (body weight >25 kg) Atenolola Oral 1 mg/kg per 1–2 mg/kg/day 1 Tablets 25, 50, 100 mg dose Maximum 100 mg/day or Syrup 25 mg in 5 mL 50 mg/day when GFR Caution in asthma although <10 mL/min/1.73 m2 cardioselective Enalapril Oral 100 µg/kg 200–500 µg/kg in a day 1 Tablets 2.5 mg, 5 mg,10 mg per dose Maximum 600 µg/kg in a Commencing treatment in day or 40 mg/day pregnancy and hyperkalaemia contraindicated Caution in renal artery stenosis and when GFR <30 mL/min/ 1.73 m2 Avoid in neonates – especially premature Furosemide Oral 500 µg/kg 1–4 mg/kg/day 1–4 Tablets 20 mg, 40 mg per dose Syrup 50 mg in 5 mL

a Recent studies in adults8,9 and NICE recommend that atenolol should not be used alone as first-line therapy. min D in the body to ensure strong and healthy In infants the phosphate binder may be bones. The kidneys are responsible for getting rid added to the feed but must be mixed well to of the phosphate from food. Phosphate is pres- avoid precipitation. The dose is tailored to the ent in protein-containing food, particularly dairy child’s phosphate level and can appear to be a products. In CRF the level of phosphate in the very large dose. If the child is receiving an blood rises, which causes increased production overnight feed a proportionately larger dose of of the parathyroid hormone (PTH). An excessive the phosphate binder may be added to that feed. level of PTH causes damage to the bones, the In kidney failure calcium is lost from the bone marrow and the lining of the blood vessels. bones and thus the body so the calcium level The amount of phosphate in the diet should needs to be maintained. Some calcium may be therefore be controlled. Treatment is started if derived from the calcium in the phosphate the plasma phosphate is above 1.7 mmol/L or binder but often a vitamin D preparation such above 2 mmol/L in neonates. A phosphate as alfacalcidol will also be prescribed and this binder such as calcium carbonate, (licensed as will raise the serum calcium by increasing Calcichew, Adcal, unlicensed as dispersible gastrointestinal calcium absorption. Alfacalcidol tablets and suspension), calcium acetate (‘off which is available as a liquid in addition to cap- label’) or, if the calcium level is high, magnesium sules should only be prescribed in nanograms or carbonate (unlicensed) or sevelamer (‘off label’) micrograms, not millilitres. is given with food to bind the dietary phosphate Even if blood calcium and phosphate levels in the gut and stop its absorption. are brought under control, high PTH levels can Monitoring and treatment of children 167

Table 13.3 Antihypertensive agents for infants less than 1 year old

Drug Route Normal Normal and Divided Preparations starting maximum doses in and comments dose dose range a day

Captopril Oral 50 µg/kg 0.5–3 mg/kg/day 3 Tablets 2 mg, 12.5 mg, 25 mg per dose Maximum 6 mg/kg/day Very soluble in water Caution in renal artery stenosis, hyperkalaemia and when GFR <30 mL/min/1.73 m2 Test dose required Propranolol Oral 1 mg/kg 1–4 mg/kg/day 3 Syrup 50 mg in 5 mL per dose Maximum 8 mg/kg/day Contraindicated in asthma and heart failure When GFR <20 mL/min/ 1.73 m2 start at lower dose

Various reference sources used: Great Ormond Street Hospital, Guy’s and St.Thomas’ Formulary etc.

also be caused by the lack of the active form of blood falls. Erythropoietin is usually given as a vitamin D. Vitamin D from food (and sunlight) subcutaneous injection once a week, or intra- needs to be activated by the kidney, so the more venously three times a week if on haemo- usable form alfacalcidol is given. dialysis. Iron also needs to be given, either Renal bone disease begins very early in orally or intravenously. kidney failure so it is important that the blood The National Institute for Health and PTH is measured regularly, in addition to the Clinical Excellence (NICE) has produced a clin- frequent monitoring of calcium and phos- ical practice guideline on ‘Anaemia manage- phate. The PTH should be in the range ment in people with chronic kidney disease’.7 0.7–5.6 pmol/L. The guidance states that for children aged 2 years and older, haemoglobin range should be maintained at the target range for adults (i.e. Anaemia (haemoglobin and iron levels) between 10.5 and 12.5 g/dL), and for children aged under 2 years treatment should maintain The kidney is responsible for making the hor- stable haemoglobin levels between 10 and mone erythropoietin, necessary for making red 12 g/dL (Table 13.4). blood cells. Children with CRF often become Use intravenous iron if persistently low anaemic and the level of haemoglobin in the ferritin levels are recorded (<100 µg/L) despite

Table 13.4 Evaluation of anaemia as used at Great Ormond Street Hospital, London

Age Normal (standard deviation) Evaluate for anaemia when range for haemoglobin (g/dL) the haemoglobin falls to (g/dL)

< 6 months 11.5 (9.5–13.5) <10 6–24 months 12.0 (10.5–13.5) <11 By 12 years of age 13.5 (11.5–15.5) <12 168 Chapter 13 • Paediatric kidney disease

globin within six months of being seen by a Table 13.5 Target haemoglobin levels paediatric nephrologist, unless there is a specific reason (Table 13.5). Age Target haemoglobin (g/dL) All children should achieve a serum ferritin <6 months > 9.5 of >100 and <800 µg/L whether or not they are 6–24 months >10 receiving epoetin. The NICE clinical guideline >2 years >10.5 contains an overview outlining the key stages of managing anaemia in chronic kidney disease (CKD).7 oral iron. Haemoglobin should be measured Figure 13.1 describes the management of once or twice a month and iron status every renal anaemia in children at Great Ormond three months. Street Hospital, including commencement of Children should achieve their target haemo- erythropoietin therapy.

Low Hb

Start oral iron if ferritin < 100 µg/L at a dose of 6mg/kg elemental Fe2+ per day

Check vitamin B12 and folate

Erythropoietin 100 units/kg/week subcutaneously Rarely, 50 units/kg three times a week IV if on haemodialysis

Measure Hb and reticulocytes weekly Ferritin every two weeks Transferrin saturation if poor response

If Hb response < 1g/dL, consider

Functional iron deficiency; Occult infection, blood loss, low B12 or folate, if transferrin saturation < 20% OR haemoglobinopathy, or inadequate dialysis

Give IV iron

If no cause found, increase dose of erythropoietin by 25 units/kg/week

Figure 13.1 Protocol for the commencement of erythropoietin as used at Great Ormond Street Hospital, London. Transplantation 169

require bilateral nephrectomies and/or a period Table 13.6 Dose of folic acid of dialysis (e.g. in focal and segmental glomer- ulosclerosis). Transplantation in the child is Age Dose often live-related (i.e. the kidney is taken from Infants 250 µg/kg once daily a living relative) so can be pre-emptive. Children 1–5 years 2.5 mg daily Pre-transplantation all vaccines must be up Children >5years 5 mg daily to date (including BCG, hepatitis B, varicella). Glandular fever (EBV) vaccine may be available soon. An accelerated immunisation procedure Folic acid may be carried out so that the child is ready for transplantation by the age of 19–24 months Folic acid is also needed to make red blood cells (Tables 13.7 and 13.8). but also seems to have a protective effect on the At Great Ormond Street Hospital, the follow- heart and blood vessels. The dosage used in chil- ing guidelines on immunisation for infants and dren is shown in Table 13.6. children with CRF are used:

• All children must complete all routine child- Potassium hood vaccines. • BCG, varicella, pneumococcal and hepatitis B Progressive renal failure can be associated with vaccines must be added in children potassium retention. Hyperkalaemia generally approaching dialysis and transplantation. does not occur until GFR is <10% of normal, • Immunisations must begin as soon as poss- however regular biochemical assessment must ible in the child born with severe CRF be carried out in all patients. Possible causes of (including infants with congenital nephrotic hyperkalaemia include: inadequate energy syndrome) as the schedule can rarely be com- intake; medicines such as the ACE inhibitors or pleted before 18 months of age. the potassium-sparing diuretic spironolactone; a • Only in exceptional circumstances can trans- high dietary intake of potassium; constipation. plantation occur without completing the full schedule.

Important general points: Transplantation • Routine immunisation in the first year of life is with the following vaccines: diphtheria, Ideally transplantation should be carried out tetanus, acellular pertussis (DTaP), inactiv- before dialysis for the following reasons: ated polio, Haemophilus influenzae type b conjugate (Hib) (Pediacel), pneumococcal • A successful transplant is the best treatment vaccine (Prevenar) and meningococcal group for children as it makes their lives as normal C conjugate. as possible. • Before school or nursery school entrance (but • Long-term mortality is lower with a trans- preferably one year after completing the plant than on dialysis. primary course) a booster of diphtheria, • The risk of calcium deposits in blood vessels tetanus, acellular pertussis and inactivated with long-term blood vessel damage is high polio should be given. on dialysis. • Measles, mumps and rubella (MMR) vaccina- • It is important to preserve access points for tion may be given at 13 and 16 months in dialysis (arm and leg veins for haemodialysis, the infant in or approaching end stage renal peritoneum for peritoneal dialysis) for future failure (ESRF). use. • Over 10 years of age give a booster dose of Transplantation without dialysis may not low-dose diphtheria, tetanus and inactivated always be possible in some conditions that polio and MMR. 170 Chapter 13 • Paediatric kidney disease Given each autumn for at--risk patients >6 months old. IM if  booster required SC ) as used at Great Ormond Street Hospital, 2 MMR(measles, zoster Varicella- Influenza vaccine rubella) Deep SC  If not predicted for ESRF can have 2nd dose with pre- school booster  b (0.5 mL) (0.5 mL) (0.5 mL) IM    Booster doses may be required months proceed to transplant. polio (Infanrix-IPV, Repevax). polio (Infanrix-IPV,    Deep SC or IM    If not previously immunised: 12 months–5 years: 2 doses separated by 2 months interval >5 years: conjugate vaccine is NOT given conjugated conjugate mumps and vaccine (Hib no DTaP/IPV) Haemophilus influenzae polio (IPV) (Pediacel) (Prevenar) type b (Hib) and inactivated vaccine Deep SC or IM IM or IM     If not previously immunised: 1–10 years give 1 dose of Hib (but for full cover should have had 3 doses of Pediacel) a Immunisation schedule for infants and children with chronic renal failure (GFR <60 mL/min/1.73 m  Or at any time up to 6 years of age with no skin test (Mantoux) required, unless born in or visited (>1 month) a high-incidence country Over the age of 6 years only if negative tuberculin skin test (Mantoux); delay transplant for 3 months Table 13.7 Table London 0 2 3 4 6 7 For notes on intradermal dose see Table 13.8. For notes on accelerated immunisation schedule see Table 13.8. 12 13 16 18 (months)(Can be (Statens Serum acellular pertussus (DTaP), Pneumococcal group C varied Institut) Age BCG SSI Diphtheria, tetanus, 7-valent Meningococcal Hepatitis B a b Over 10 years of age: Booster dose of low-dose diphtheria, tetanus and inactivated polio (Revaxis). If schedule followed at 19 After second single birthday, dose of 23-valent pneumococcal polysaccharide vaccine (Pneumovax II). Before school or nursery school entry if not ‘on call’: Booster doses of diphtheria, tetanus, acellular pertussis, inactivated Transplantation 171

Table 13.8 Notes on immunisation schedule for infants and children with chronic renal failure

Vaccine Comments

BCG SSI (Statens Serum Institut) May be given to some newborns routinely Can be given up to the age of 6 years without a prior Mantoux test, unless born in or visited (>1 month) a high-incidence country. Over the age of 6 years give only if tuberculin skin test (Mantoux) negative (NB Tuberculin testing should not be carried out within 3 weeks of receiving a live vaccine as response may be falsely negative) Age <12 months BCG dose 0.05 mL by intradermal injection Age >12 months BCG dose 0.1 mL by intradermal injection May be given simultaneously with another live vaccine but if not given at the same time, allow an interval of at least 3 weeks In neonates the vaccine must be given intradermally into the upper arm only (preferably the left deltoid region). Do not use the same arm for further immunisation for 3 months Delay transplantation for 3 months after BCG Hepatitis B All patients who will need ESRF management should be immunised against hepatitis B, preferably pre-emptively while the GFR remains relatively high Can be given at any age at intervals of 0, 1 and 6 months An accelerated course can be used so that the third dose is given 2 months after the first dose (i.e. doses at 0, 1 and 3 months and a booster dose at 12 months) For IM administration: The anterolateral thigh (IM) is the preferred site in infants and young children. The deltoid muscle is the preferred site in older children. It should not be injected into the buttock as vaccine efficacy is reduced Dose for pre-dialysis patients: Engerix B (GlaxoSmithKline) by IM injection Birth to 10 years 3 doses of 0.5 mL (10 µg) Age 10–15 years 3 doses of 1 mL (20 µg) HB-Vax Pro (Aventis Pasteur ) by IM injection Under 10 years 3 doses of 0.5 mL (5 µg) Age 10–15 years increase dose to 1 mL (10 µg) These doses should be doubled for patients on dialysis and 4 doses given at 0, 1, 2 and 6 months Check anti HBsAg antibodies 2–3 months after the third dose Dialysis patients should be monitored annually and revaccinated if necessary Antibodies 100 iu/L Protective – give booster every 5 years 10–100 iu/L Poor responder – give booster at 1 and 5 years <10 iu/L Non-responder – repeat course of vaccine MMR Give at 13 months (0.5 mL by deep SC or by IM injection). If ESRF not imminent, 2nd MMR can be given with pre-school booster. If in ESRF, 2nd MMR can be given at 16 months of age (interval of at least 3 months after 1st vaccine) (continued overleaf) 172 Chapter 13 • Paediatric kidney disease

Table 13.8 (continued)

Vaccine Comments

MMR (continued) Children <4 years old with a GFR <30 mL/min/1.73 m2 should have their pre-school/nursery school booster (2nd MMR) brought forward. Older children – a 2nd MMR is advised unless there is definite serological evidence of immunity. If administering at the same time as other injections, use a separate syringe and needle; give MMR first as it is less painful, and use a different limb. Alternatively, a second appointment can be made. Other live vaccines may either be given on the same day or at least 3 weeks later Check measles antibody response 2–4 weeks after completing MMR course An initial negative or equivocal antibody result should be repeated. In vaccinated children who have had a previously positive measles IgG but are found to be negative/equivocal on retesting, the conventional wisdom is that primed memory cells will respond to a measles challenge. However, if blood or blood products have been given 3 months prior to the test, the measles IgG may be transiently positive and a retest will be required Delay transplantation for 1 month after MMR course Varicella vaccine (Varilrix or Can be given with, or 4 weeks after MMR vaccine if non-immune (check titres). Varivax) Ensure lymphocyte count >1.2 109/L Delay for 3 months if patient has received immunoglobulin or a blood transfusion because of likelihood of vaccine failure due to passively acquired varicella antibodies Salicylates should be avoided for 6 weeks after varicella vaccination as Reye’s syndrome has been reported following the use of salicylates during natural varicella infection In ‘healthy’ patients if a measles containing vaccine is not given at the same time as the varicella vaccine an interval of at least one month must elapse between vaccines. Measles vaccination may lead to short-lived suppression of Dose: the cell mediated response Age >12 months–12 years 1 dose (0.5 mL) SC (upper arm-deltoid region). May need a booster dose after an interval of 2 months Age >13 years 2 doses with an interval between doses of 4–8 weeks In high-risk patients additional doses might be required Check titres (ELISA) after 2–3 months. If no seroconversion a second dose is given Delay transplantation for 1 month after vaccination course if seroconversion demonstrated Pneumococcal vaccines Saccharide conjugated vaccine-7- Previously recommended for immunosuppressed children aged 2 months to 5 valent (Prevenar) years or who have CRF and/or nephrotic syndrome, although now routine in the UK at 2 and 4 months (with DTP/IPV and Hib) and at 13 months (with MMR) For previously unvaccinated older infants and children: Age 6 months–12 months 2 doses (0.5 mL) at least a month apart. A third dose is recommended after the 1st birthday Age 12 months to 5 years 2 doses (0.5 mL) with an interval of at least 2 months between doses The vaccine is not necessary in children over 5 years of age → Dialysis 173

Table 13.8 (continued)

Vaccine Comments

Pneumococcal vaccines (continued) Pneumococcal polysaccharide vaccine-23-valent (Pneumovax II) All children between 2 and 5 who have received the pneumococcal saccharide vaccine need a single dose years (0.5 mL) to provide protection against the serotypes of Str. pneumoniae not covered in the conjugate vaccine Leave an interval of at least 2 months between the two vaccines Children >5 years need a single dose Children >10 years can be revaccinated if high risk (e.g. nephrotic syndrome) Influenza vaccine Annual vaccination is recommended from the age of 6 months

For up to date information refer to the care professionals prescribing these medicines Department of Health website at www.doh.gov. should ensure that children and adolescents uk and to the vaccine manufacturer’s literature. receiving renal transplants and/or their legal NICE has recently published its final guardians are aware of this, and that they appraisal determination on immunosuppressive consent to the use of these medicines in these therapy for renal transplantation in children circumstances.’10 and adolescents.10 Between April 2003 and Each paediatric unit will have its own renal March 2004 approximately 130 patients (7% of transplant protocol, which will be continually those who underwent renal transplantation) subject to trials of combinations of drugs to were under 18 years of age. The document optimise the preservation of the graft with min- recognises that organ transplantation is not imal side-effects. In children the use of steroids considered fully successful for children and and their effect on linear growth is a major adolescents unless they grow and develop as factor in considering the rapid reduction of normal after transplantation. Concordance due steroids or steroid-free immunosuppressant to complex medication regimens and/or associ- regimens. As the regimens are complex they will ated side-effects may be a problem. The docu- not be discussed here. ment considers the use of: the induction agents Patients are continually monitored for infec- basiliximab and daclizumab; the calcineurin tion, including viral infections and opportun- inhibitors ciclosporin and tacrolimus; and aza- istic protozoal, fungal and bacterial infections, thioprine and corticosteroids. The use of post-transplant lymphoproliferative disorders, mycophenolate mofetil in corticosteroid reduc- de novo post-transplant diabetes mellitus and tion or withdrawal strategies is recommended evidence of rejection. only within the context of randomised clinical trials, mycophenolate sodium is currently not recommended and sirolimus is only recom- Dialysis mended when proven intolerance to calcineurin inhibitors (including nephrotoxicity) neces- sitates the complete withdrawal of these Waste products are cleared from the blood by treatments. diffusion. The strong solution, blood containing The document also recognises that some creatinine and urea, passes to the weaker solu- medicines may be prescribed outside the terms tion, the dialysate. Excess water is removed by of their UK marketing authorisation. ‘Health- ultrafiltration. In peritoneal dialysis glucose 174 Chapter 13 • Paediatric kidney disease from the dialysate draws water from the blood; Peritonitis associated with dialysis in haemodialysis the machine draws water from the blood. Waste and water pass into the dialysis If infection enters into the abdomen, peritonitis fluid (dialysate) for removal from the body. The will develop. The dialysate, which is usually dialysis membrane keeps the dialysis fluid and clear becomes cloudy and the child develops blood apart. In peritoneal dialysis the periton- abdominal pain and may have a temperature. eum (layer of cells that lines the abdomen and Peritonitis may be treated by giving antibiotics covers the guts) is the membrane; in haemo- intravenously or added to the dialysate. The dialysis a filter is used. protocol used at Great Ormond Street Hospital is shown in Table 13.9. Review after 24–48 hours with blood cul- Peritoneal dialysis ture and peritoneal dialysis fluid (PDF) results. If blood culture is negative, change to intra- This can be undertaken at home so that the peritoneal (IP) treatment as above. If blood child can go to school. A soft catheter culture is positive, continue IV antibiotics (Tenckhoff) is tunnelled under the skin into the according to sensitivities, and consider adding abdomen, the tip being in the pelvis. To allow IP antibiotics. the incision to heal properly and prevent any Continuing antibiotic therapy will be modi- risks of infection dialysis is not usually started fied according to the identity and sensitivity of for 3–4 weeks if possible. Constipation may dis- the organisms cultured. It is essential to liaise place the tube so to avoid this lactulose is given. with medical microbiology. It may be helpful to consider substituting flucloxacillin 50 mg/L for sensitive Gram-positive organisms if there is Continuous ambulatory peritoneal dialysis concern about the development of resistance to In continuous ambulatory peritoneal dialysis vancomycin, and changing to the use of gen- (CAPD), the bag is hung and allowed to run into tamicin 5 mg/L or amikacin 25 mg/L for sensit- the abdomen. The fluid is then left in the ive Gram-negative organisms if ciprofloxacin abdomen until the next time when the old fluid resistance is a concern. When no bacteria are will be drained out and replaced with fresh isolated both antibiotics are continued. fluid. This is done four times a day: morning, lunchtime, late afternoon and before bed. It Dialysis regimen for peritonitis may be better for older children who want to be able to go out in the evenings. • All children should initially be treated with CCPD (home choice) using usual TTV (total therapy volume) but with a TTT (total ther- Continuous cycling peritoneal dialysis apy time) of 24 hours. This will give the same Also called automated peritoneal dialysis (APD), number of cycles per day but with increased in continuous cycling peritoneal dialysis dwell times and should be continued for (CCPD) a machine, rather than gravity, is used 2 days (48 hours). Antibiotics and heparin to push the fluid in and out of the abdomen. should be added to all of the dialysis fluid Dialysis takes place at night whilst the child is bags if applicable (heparin only if on IV asleep. Usually 10–12 hours on the machine antibiotics). is needed so this is better for younger chil- • Close observations of fluid balance and dren. Sometimes an extra cycle can be put in plasma potassium levels should be performed. during the day so that the length of time • If the white blood cell count (WBC) is overnight can be shortened in the older chil- <100 106/L after 48 hours, continue on dren. Usually some fluid is left in during the usual dialysis regimen adding antibiotics to day. Initial dialysis fluid volumes are 10 mL/kg the PDF as above (including last fill if on gradually increasing up to 45–50 mL/kg (or CCPD). If the WBC is >100 106/L after 48 1100–1400 mL/m2). hours continue cycling regimen. Dialysis 175

Table 13.9 Protocol for treatment and prevention of peritonitis in children on peritoneal dialysis as used at Great Ormond Street Hospital, London

Clinical presentation Cloudy dialysate fluid ± Abdominal pain ± Fever ± History of line break/contamination Assessment Either local infection with minor systemic signs or associated severe systemic illness. Send sample of PDF effluent for cell count and differential, Gram stain & culture (MC&S). PDF sample should be 50–100 mL, with the dwell time and fill volume indicated on the form Perform a CRP, WBC and differential blood cultures if clinically indicated Diagnosis WBC >100 106/L ± Organisms on Gram stain (Gram positive / negative or yeasts) ± Positive culture Note: If WBC 50–100 106/L and patient’s symptoms/signs suggestive of peritonitis, initiate treatment If WBC 50–100 106/L and patient asymptomatic, hold dialysis and repeat specimen in 4–6 hours Treatment with antibiotics If local signs/minor systemic Initially use both IP vancomycin and ciprofloxacin until Gram stain/culture of illness: PDF available. Then modify treatment accordingly Gram-positive organisms Gram-negative organisms Vancomycin (15 mg/L) IP No organisms seen but Ciprofloxacin (20 mg/L) IP WBC >100 106/L Vancomycin (15 mg/L) IP + ciprofloxacin (20 mg/L) IP + heparin 200 units/L (for 48 hours cycling and continue as clinically indicated) + nystatin oral suspension 100 000 units four times a day whilst on antibiotics If severe systemic illness: Initially use both IV vancomycin and ciprofloxacin until Gram stain/culture of PDF available. Then modify treatment accordingly Consider this approach with immunocompromised patient Gram-positive organisms Vancomycin 10 mg/kg stat IV (if 24-hour level <10 mg/L give further dose) Gram-negative organisms Ciprofloxacin 5 mg/kg IV 12 hourly (max 400 mg 12 hourly) No organisms seen but Vancomycin 10 mg/kg stat IV (if 24-hour level <10 mg/L give further dose) WBC >100 106/L + ciprofloxacin 5 mg/kg dose 12 hourly IV (max dose 400 mg 12 hourly) + heparin 200 units/L IP (for 48 hours cycling and continue as clinically indicated) + nystatin oral suspension 100 000 units four times a day whilst on antibiotics

• Duration of treatment: 14 days (may require persistent cloudy effluent. If >10% leucocytes up to four weeks for Staph. aureus). are eosinophils this is suggestive of • Send daily PDF samples to microbiology to eosinophilic peritonitis. May also have a monitor treatment response. peripheral blood eosinophilia. No extra treat- • Send sample for eosinophil count to haem- ment indicated. atology if culture-negative peritonitis and • In most cases the patient can go home after 176 Chapter 13 • Paediatric kidney disease

48 hours to continue treatment. The com- Frequent episodes of peritonitis can damage munity team is involved if parents cannot the peritoneum and reduce the length of time add antibiotics to bags. that peritoneal dialysis can be given. On aver- • Remove catheter if the following develop: age, peritoneal dialysis can be used for 5 years fungal peritonitis; severe intra-abdominal before it ceases to work properly. sepsis and septicaemic shock; exit site or tunnel infection due to the same organism as the peritonitis (recurrence within four weeks Haemodialysis with the same organism); no decline or increase in dialysate white cell count after For haemodialysis there needs to be access 3–4 days if infection severe or 7 days if infec- to the blood vessels so that blood can be taken tion mild. out of the body, passed through a filter to clean • Consider removal of catheter if child remains it, and then returned to the body. Access is symptomatic after 3–4 days. via a catheter or by a fistula. Haemodialysis takes place for 4 hours on 3 days per week. Post-surgical peritonitis (within two weeks of Schooling is arranged whilst the child is on procedure) haemodialysis. The catheter used for haemodialysis needs to A raised PDF white cell count is often found fol- be relatively large to allow high blood flow lowing an intraperitoneal procedure (i.e. place- speeds – a Permcath consisting of two lumens ment of peritoneal dialysis catheter) but (one for blood to leave the body and one for symptomatic peritonitis is uncommon. The return) is placed in the vessels in the neck. operative procedure is covered with IV anti- Permcaths are only used in an emergency or if biotics (amikacin 10 mg/kg and teicoplanin haemodialysis is short term or in young chil- 10 mg/kg stat) and the catheter is capped off fol- dren as they easily become infected and may lowing frequent flushes until the dialysate is damage the blood vessel. If the catheter clear. The catheter is flushed weekly but a PDF becomes infected, vancomycin or teicoplanin sample should only be sent to microbiology if ‘line-locks’ may be used according to the sensit- the child is symptomatic. Treatment will be ivities of the infecting organism. indicated in a symptomatic child with a rising Older children are dialysed via a fistula. An WBC on serial PDF samples. artery is joined to a vein in the arm; this makes the vein increase in size and become tougher so Line break/contamination that needles can be placed into the vein and If a line break/contamination occurs when peri- high blood speeds through the filter can be toneal dialysis has commenced: obtained. A local anaesthetic cream is used before the needles are put in. The vessels in the • Perform a line change and obtain PDF sam- neck are preserved and the infection risk is ple as before. reduced. After creation the fistula will take 4–6 6 • If WBC is <100 10 /L add vancomycin and weeks before it is usable. ciprofloxacin (as above) to dialysis bags for Paediatric haemodialysis brings another set 48 hours but continue usual dialysis regimen. of challenges and requires specialist knowledge 6 • If WBC is >100 10 /L then treat as peri- and expertise. tonitis (as above). If line break/contamination occurs before peri- toneal dialysis has commenced: Fluid restriction on dialysis • Perform a line change. Give IV antibiotics for This is dependent upon how much urine is 48 hours and continue as clinically indicated. passed (a small amount is lost in stools and A summary of the treatment of peritonitis in sweat) and restriction will depend upon how peritoneal dialysis is given in Table 13.10. well fluid is removed with peritoneal dialysis, Dialysis 177

Table 13.10 Treatment of peritonitis

Treatment

Recurrent peritonitis Consider risk factors: Revisit exchange technique Tenckhoff catheter exit site or Swab exit site and obtain ultrasound scan of tunnel. May need further course of tunnel infection oral antibiotics (up to 6 weeks) according to sensitivities. Infected superficial cuff can be exteriorised and shaved. If inner cuff of catheter involved will need catheter replacement Staph. aureus infection Check if child or carers are nasal carriers. If positive nasal swabs, apply topical nasal mupirocin twice a day Gastrostomy exit site If appears infected obtain swab and treat appropriately

Note: Replace peritoneal dialysis catheter if refractory peritonitis (i.e. relapsing or >2 recurrent infections) as possible biofilm formation

Fungal peritonitis Children at risk: Frequent broad-spectrum Start treatment with liposomal amphotericin – 1 mg/kg as a daily dose IV, antibiotic usage changing if possible after 48 hours (following fluconazole sensitivity testing and identification) to fluconazole 12 mg/kg IV as a single daily dose for 48 hours decreasing to 6 mg/kg/ day (max dose 200 mg daily) for a total of at least 2 weeks If functioning renal transplant, adjust dose based on renal function Then continue with oral fluconazole for further 4 weeks. Most Candida albicans (germ tube-positive yeast) is sensitive to fluconazole Immunosuppressed post Catheter removal as soon as possible. transplant with peritoneal dialysis catheter in situ Gastrostomy (but no significant relationship)2

Note: Re-initiation of peritoneal dialysis is common following successful treatment of fungal peritonitis.11,12

and how much fluid the child can tolerate being protein is lost in the dialysate. Some sugar will taken off with haemodialysis in a relatively be absorbed from the dialysate. short time. On haemodialysis, if too much is drunk On peritoneal dialysis, what is taken in must between dialysis sessions the blood pressure equal what comes out if the child is not to goes up and the child can become very unwell, retain fluid and gain weight (oedema). Higher particularly during the process of fluid removal. concentrations of dialysate will pull off more The child will also need to stay on the machine fluid. In the infant and young child a lot of longer. On haemodialysis, potassium must be adjustment is required with dialysis concentra- monitored carefully. tions, types of fluid and fill volumes to optimise Neither type of dialysis is good at removing growth (not oedema) and development. The phosphate so a phosphate binder is essential. amount of protein in the diet is increased as High levels of phosphate and PTH are important 178 Chapter 13 • Paediatric kidney disease

Table 13.11 Intravenous iron dose in haemodialysis

Maintenance dose

Ferritin <100 µg/L and TSAT <20% A 7 mg/kg (max 200 mg) IV stat dose is given, followed by 2 mg/kg/dose IV every 2 weeks (max single dose 100 mg)

Ferritin >100 and <800 µg/L, TSAT 20% 2 mg/kg/dose IV every 2 weeks (max single dose 100 mg)

Ferritin >800 µg/L, TSAT >50% No treatment required

The serum ferritin (µg/L), iron (µmol/L) and transferrin (µmol/L) should be measured monthly. Transferrin saturation (TSAT) can be calculated as follows: TSAT = iron/2 transferrin (%). From ref. 13. causes of cardiovascular disease. The mortality The gastric pH is alkaline at birth. Gastric is about 700 times higher in young adult acid secretion begins within a day or two and patients on long-term dialysis than in the adult levels are reached at age 5–12 years. normal population. Intestinal enzyme activities affecting drug Erythropoietin and iron needed before dia- absorption may be subject to age-dependent lysis will need to be continued, but iron is alterations. Protein binding is altered by usually given intravenously during haemo- acidosis, malnutrition and inflammation. The dialysis as iron sucrose (off-label) at the doses extent to which drugs are cleared by haemo- shown in Table 13.11. dialysis or peritoneal dialysis is also important. The risk of death in children on dialysis is Drug action can also interfere with a child’s about 1% per year. development (e.g. long-term corticosteroids can inhibit growth and cause osteoporosis). To date it is impossible to accurately deter- mine all factors that may influence drug meta- Drug prescribing in renal disease bolism in children in every paediatric age group. Thus continual observation and wherever poss- Compared with adult patients there are few ible careful monitoring of drug levels is essen- published studies on drug dosing for children tial, particularly in those with a narrow with renal impairment. Age, weight or body sur- therapeutic range. face area needs to be considered in addition to References such as the British National renal function. Dosage based on body weight is Formulary for Children, Guys & St Thomas often inappropriate and may result in under- Paediatric Formulary or Daschner’s dosing dosing in small infants. Adjusting the dose to tables3 may be used. body surface area gives a better estimate of the The Great Ormond Street protocols listed in dose for an infant or child. Surface area this chapter are those in current use but are increases at a lower rate than body weight dur- regularly subject to review and may change. ing a child’s growth and many physiological parameters that are important for drug distribu- tion and elimination are closely correlated to Acknowledgements surface area. Immature liver and kidney func- tions in newborns markedly improve during the To all the staff in the nephrology unit at Great first months of life, altering drug metabolism. Ormond Street Hospital, London and to Kuan Changes in gastrointestinal motility, gastric pH, Ooi, pharmacy department, for assistance in nausea and vomiting all alter drug absorption. proofreading and formatting. Gut motility and gastric emptying time are delayed in neonates. References 179

C CASE STUDY

A 1-day-old term baby (weight 3 kg) is admitted in chronic renal failure with a diagnosis of bi- lateral dysplastic kidneys. Urea and electrolytes are as follows (normal ranges within parentheses): • Sodium 136 mmol/L (133–146 mmol/L) • Potassium 3.9 mmol/L (3.2–6.0 mmol/L)

• Total CO2 23 mmol/L (17–27 mmol/L) • Urea 5.0 mmol/L (0.7–5.0 mmol/L) • Creatinine 234 µmol/L (53–97 µmol/L) • Calcium 2.27 mmol/L (1.96–2.66 mmol/L) • Magnesium 0.78 mmol/L (0.66–1.0 mmol/L) • Phosphate 2.38 mmol/L (1.5–2.6 mmol/L) • Albumin 29 g/L (26–36 g/L) • PTH 10.0 pmol/L (0.7–5.6 pmol/L) • Liver function tests within normal range • Haemoglobin 12 g/dL (13.5–19.5 g/dL).

Q1. How would you determine the baby’s GFR?

Q2. Describe the therapy that should be initiated and why.

Q3. How would you ensure that the child maintains optimum growth and development?

Q4. The parents are told that the baby will need to start dialysis in the very near future, and will ultimately require a transplant. The parents are very keen to donate a kidney as soon as possible. They are aware of the work up required, but what is the earliest age that a transplant could go ahead and why? A peritoneal dialysis catheter is inserted and peritoneal dialysis started. Once dialysis is stabilised and after extensive training, mum feels confident to perform the dialysis at home. The baby is dis- charged. However 2 days later mum phones the unit to say that her baby was screaming and had a temperature. She had also noticed that the effluent dialysis bag was cloudy.

Q5. What is the likely diagnosis and what would you therefore recommend that mum should do?

References inine formula in the measurement of glomerular filtration rate. Arch Dis Child 1982; 57: 611–615. 3. Daschner M. Drug dosage in children with 1. Department of Health. Medicines standard: reduced renal function. Pediatr Nephrol 2005; 20: National Service Framework for Children, Young 1675–1686. People and Maternity Services. October 2004. 4. Mattman A, Eintracht S, Mock T et al. Estimating http://www.dh.gov.uk pediatric glomerular filtration rates in the era of 2. Morris MC, Allanby CW, Toseland P, Haycock GB, chronic kidney disease staging. J Am Soc Nephrol Chantler C. Evaluation of a height/plasma creat- 2006; 17: 487–496. 180 Chapter 13 • Paediatric kidney disease

5. Fawer CL, Torrado A, Guignard JP. Maturation of tation in children and adolescents. 2006. http:// renal function in full term and premature www.nice.org.uk neonates. Helv Paediatr Acta 1979; 34: 11–21. 11. The Mupirocin Study Group. Nasal mupirocin 6. National High Blood Pressure Education Program. prevents Staphylococcus aureus exit site infection Working Group on high blood pressure in chil- during peritoneal dialysis. J Am Soc Nephrol 1996; dren and adolescents. Pediatrics. 2004; 114 (Suppl 7: 2403–2408. 4th report): 555–576. http://pediatrics.aappubli- 12. Warady B, Bashir M, Donaldson L. Fungal peri- cations.org/cgi/reprint/114/2/S2/555 tonitis in children receiving PD: A report of the 7. National Institute for Health and Clinical NAPRTCS. Kidney Int 2000; 58: 384–389. Excellence (NICE). Clinical guideline 39: 13. Morgan HEG, Gautam M, Geary DF. Maintenance Anaemia management in chronic kidney disease: intravenous iron therapy in pediatric haemo- Understanding NICE guidance. September 2006. dialysis patients. Pediatr Nephrol 2001; 16: http://guidance.nice.org.uk/cg39/publicinfo/pdf/ 779–784. English 8. Carlberg B, Samuelsson O, Lindholm LH. Atenolol in hypertension: is it a wise choice? Lancet 2004; 364: 1684–1689. Further reading 9. Anglo Scandinavian Cardiac Outcomes Trial (ASCOT) study aiming to evaluate different treat- ment strategies to prevent cardiovascular disease Stein A, Wild J. Kidney Failure Explained, 2nd edn. in hypertensive patients. http://www.ascotstudy. London: Class Publishing, 2002. [Although this org book is about adults living with kidney disease it 10. National Institute for Health and Clinical is very well written and presented.] Excellence (NICE). Final appraisal determination: Rees L, Brogan P, Webb N. Handbook of Paediatric Immunosuppressive therapy for renal transplan- Nephrology. Oxford: Oxford University Press, 2006. 14

Renal pharmacy in critical care

John Dade

A large proportion of patients admitted to an understanding of the following issues amongst intensive care unit (ICU) will experience some others: reversible changes in their renal function during • The causes of renal impairment in critically the course of their admission as a consequence ill patients and how this can be prevented. of their critical illness. A smaller and significant This is a key role bearing in mind the mor- number will develop acute renal failure (ARF). tality rates once ARF occurs. This chapter will concentrate on the pharma- • How renal replacement techniques are per- ceutical needs of patients who develop ARF and formed, particularly continuous haemofiltra- require an ICU admission. It will also briefly tion and continuous haemodialysis. review the use of drug therapies, specific to the • How to adjust drug therapy to allow for the critical care, in patients with renal dysfunction. effects of renal impairment and replacement Acute renal failure is a common problem in (e.g. dosage, fluid allowance, adverse events the intensive care unit and affects approxim- and contraindications). ately 10 000 patients in the UK annually.1 This equates to 5–20% of ICU admissions, depending on the individual ICU’s patient mix. Lower rates are seen in single speciality units (e.g. cardio- Preventing acute renal failure thoracic ICUs), and higher rates in general ICUs. Acute renal failure is a serious medical con- dition. Even if you allow for the severity of their Acute renal impairment and ARF most com- concurrent illness, patients with ARF have a sig- monly occurs following a period of under- nificantly worse outcome; patients receiving perfusion of the kidney (pre-renal failure), ventilation and renal replacement have an ICU typically as a result of hypotension and/or mortality rate in excess of 50%, whereas on hypovolaemia. The causes for underfilling and average 20–25% of all patients fail to survive low blood pressure can vary, but the manage- their ICU admission. Worse still, a significant ment is almost always the same (i.e. administer number of individuals that recover to leave ICU fluids, and where necessary support the blood will die before leaving hospital, resulting in a pressure). Pharmacists can play a key role in total hospital mortality rate of approximately encouraging these treatments, and crucially 70% in ARF patients, compared with 35–40% reversing any drug-induced causes of hypo- for all ICU admissions. The outlook for those volaemia and hypotension. Common causes for patients who survive ARF, however, is good, hypovolaemia that should be considered with less than 10% remaining dialysis- include excessive diuretic therapy, unnecessary dependent in the longer term.1 fluid restrictions, and high gastrointestinal losses. Pharmacists who work with critically ill Causes of hypotension include administration patients will spend a significant part of their of hypotensive medications (e.g. antihyper- time managing the causes and consequences of tensive drugs and many other cardiovascular renal failure. Consequently they will require an agents) and sedative agents (e.g. propofol – a

181 182 Chapter 14 • Renal pharmacy in critical care very common cause of low BP), and opioids. See viding haemofiltration are based in hospitals also Chapter 11. with renal units, nephrologists and renal phar- Pharmacists should also be aware that sicker macy specialists on site. patients are at a higher risk of developing ARF and be vigilant to ensure that the use of nephro- toxic medications is kept to the absolute mini- Renal replacement therapies used in UK mum, particularly in the acutest phase of the intensive care units illness. Drugs to consider include non-steroidal anti-inflammatory drugs (NSAIDs), radiological There is a considerable difference between the contrast agents, aminoglycosides, amphotericin, techniques used for long-term renal support in ciclosporin, and tacrolimus. the dialysis-dependent population and those Finally the role of drug therapy in the pre- used in the ICU. A survey published in 2003 vention of renal failure is currently very limited. demonstrated that the following techniques The use of low-dose dopamine infusions, loop were used in UK ICUs:1 diuretics and mannitol has declined signifi- • Continuous veno-venous haemofiltration cantly as evidence of their lack of efficacy has (CVVHF) 88% been published. The only possible exception is • Continuous veno-venous haemodiafiltration the use of alkaline diuresis in the prevention of (CVVHDF) 70% ARF due to high myoglobin levels. This can • Intermittent haemodialysis 30% occur following significant tissue injury (e.g. • Intermittent haemofiltration 16% trauma), pressure areas or drug-induced rhabdo- • None 11%. myolysis (e.g. statins). This treatment involves the maintenance of a high output of alkalinised These findings reflect the fact that CVVHF and urine (pH >6–6.5), which is achieved by admin- to a lesser extent CVVHDF, are the mainstay istering a combination of sodium chloride 0.9%, methods used for renal replacement in ICU, par- sodium bicarbonate (1.4–8.4% according to pro- ticularly in the earlier more critically ill phase of tocol and urinary pH), and other fluids. a patient’s admission. The remaining discussion will concentrate on these techniques as they are rarely used outside of critical care areas, such as ICU high dependency units (HDUs). Inter- Renal replacement therapy in the mittent techniques are used in some ICUs but intensive care unit usually when the ICU patient is recovering from their acute illness, as their treatment becomes Up until 5–10 years ago the majority of ICUs in less invasive. See Chapter 8. district general hospitals did not have facilities for renal replacement and patients were usually transferred to tertiary ICUs for management. In Continuous renal replacement therapies the last 5 years the picture has changed consid- erably as haemofiltration equipment has The procedures of haemofiltration and become more widely available. A survey pub- haemodialysis are physiologically disruptive, lished in 2003 found that 90% of ICUs in the and regularly induce acute changes in blood UK provided renal replacement therapy in pressure, fluid balance and many other para- house, and this figure has increased to close to meters. Critically ill patients are particularly 100% in the intervening years.1 This change has sensitive to these effects, and many cannot tol- had a number of knock-on effects, the principal erate intermittent courses of renal replacement. being that in most ICUs renal replacement is As a result, continuous modes of haemofiltra- nowadays solely provided by the critical care tion and haemodialysis have been developed. team, with varying back-up support from Put simply, continuous renal replacement tech- nephrology teams. Only one-third of ICUs pro- niques are what you get if you run intermittent Renal replacement therapy in the intensive care unit 183 techniques at lower rates of clearance and blood combination of intravenous potassium flow, and over a longer, unspecified period of and/or adding potassium to the haemofiltra- time. The physical processes occurring, includ- tion fluid to a defined concentration. ing those that control drug clearance, are • Other units will use a potassium-containing identical to those occurring in intermittent haemofiltration fluid containing 2 or techniques. Refer to Chapter 8 for a fuller 4 mmol/L potassium. This is used alongside a description. potassium infusion if it is necessary to increase blood potassium levels. A potassium- free haemofiltration fluid is used if there is an Medications required during renal replacement urgent requirement to reduce blood potas- therapy sium. A number of medicines are necessary to perform Both methods have their advantages and dis- haemodialysis or haemofiltration. These are the advantages, but the latter method is more renal replacement fluid, anticoagulants and, in widely used as it is simpler to operate, and uses some ICUs, electrolytes, as these may be added smaller quantities of concentrated potassium to the replacement fluid. injection, a product whose use is increasingly being restricted in UK hospitals. Renal replacement fluid Renal replacement fluids are water-based mix- Buffers – lactate vs. bicarbonate tures of a buffer and basic electrolytes, formu- Until recently, the choice of renal replacement lated to be close to the components of an ideal buffer in critically ill patients was limited to pre- extracellular fluid. They can be presented in a cursor substances, principally lactate, and less number of ways but in most ICUs haemo- commonly acetate, that are metabolised to the filtration fluid will be used as a dialysis fluid in natural physiological buffer bicarbonate by haemodialysis, and a haemofiltration fluid the liver. These products were used for practical in haemofiltration. This fluid is the major prod- reasons as bicarbonate haemofiltration fluids are uct for controlling blood chemistry. unstable and cannot be supplied as a ready During the process of renal replacement a mixed solution. For the majority of ICU patient’s electrolyte and solute levels will gradu- patients, lactate-based fluids are well tolerated, ally move closer to the components of the as they are able to metabolise the precursors to replacement fluid (i.e. waste products and drugs bicarbonate and buffer their acidotic blood. removed, electrolytes equilibrated and buffers A small minority of patients are unable to replaced). A range of haemofiltration fluids are metabolise lactate to an adequate degree. This available, most of which have similar formula- results in worsening acidosis as there is insuffi- tions. The major differences are in the choice of cient buffer generated, and because of accumu- buffer, and the level of electrolytes, principally lation of the lactate, which is an acid in its own potassium. right. Patients at risk of this ‘lactate intolerance’ include those with significant hepatic impair- ment, and poor perfusion states secondary to Potassium severe sepsis, severe cardiac failure and systemic The optimum blood potassium for the majority inflammatory response syndrome. Lactate intol- of ICU patients will be within the range of erance becomes apparent if the acidosis fails to 4–5 mmol/L. In patients with renal failure this respond or worsens, and there are significant can be maintained in a number of ways: rises in blood lactate following the initiation of • Some units will use a potassium-free CVVHF/HDF. Until recently, managing acidosis haemofiltration as the standard fluid, and in these patients was very challenging, but this once the initial high potassium is controlled has been improved with the availability of they will maintain the serum potassium by a bicarbonate-based haemofiltration fluids. These 184 Chapter 14 • Renal pharmacy in critical care are mixed at the point of use and therefore have haemofiltration fluid flow rate will be roughly a much better shelf life. equivalent to the filtration rate of a haemo- filtration device. In most ICUs the clearance Anticoagulation rate for either device will be in the region of Both haemodialysis and haemofiltration require 25–40 mL/min, but you should check for the the use of an extracorporeal blood circuit to exact figure for accuracy and also because as a deliver blood to the artificial kidney. As a result, small number of units may use significantly some method of anticoagulation is necessary to higher flow rates. both maintain blood flow and to prevent the Once you have this figure you can start to build-up of micro-thrombi that reduce the effi- make dosing decisions. Strictly speaking, you ciency of the artificial kidney. This latter process may wish to review literature to obtain details is very important for continuous renal replace- on drug clearance but in practice pharmacists ment techniques, as filter lives need to be opti- look up the dosage recommended for this creat- mised to achieve effective control of blood inine clearance rate in the drug’s summary of chemistry.3 product characteristics (SPC) and recommend The commonest anticoagulant used in this this dosage. This method will give a reasonable context is unfractionated heparin. This is typic- dosage decision in most situations. If you have ally infused to achieve a clotting time of 2 times inadequate information to calculate dosage your normal (each hospital will use a different assay, best option is to seek guidance from a more e.g. APTT, KCTT, ACT). Unlike in most areas of experienced colleague. Finally, for drugs such as medicine, unfractionated heparin is generally aminoglycosides and vancomycin the best favoured over low-molecular-weight heparins, option is to adjust dosages according to blood because the latter is less easily monitored at the levels. bedside. If heparin is not effective, there are concerns about bleeding, or there are side-effects or con- Drug therapies specific to critical care traindications (e.g. thrombocytopenia), some areas units will use epoprostenol (prostacyclin 5 ng/kg/min) as an alternative agent. However this agent is very expensive and many units Analgesia and sedation restrict its use as much as possible. Other approaches include pre-dilution of blood with A large majority of ICU patients will receive a haemofiltration fluid, or using no anticoagula- combination of an analgesic and sedative tion if the patient’s clotting is very deranged. drug(s) during their admission. The indications Newer techniques include the use of citrate, for these drugs are principally to control post- heparinoid or hirudins, but these are rarely used operative or procedural pain, to facilitate venti- in the UK. lation, and to manage delirium and agitation. Mechanical ventilation is an uncomfortable process commonly associated with agitation. Drug dosing in patients receiving continuous renal Strong opioids are administered to control the replacement therapy pain of the process, and where necessary to A variety of strategies can be adopted when depress respiratory drive as this makes ventila- making dosage decisions in patients receiving tion easier. Sedative drugs are prescribed to continuous renal replacement therapy. The key manage agitation, to prevent the patient from information that you require is the urea clear- pulling out their endotracheal tube and lines, ance rate of the technique in use. This can be and to induce anaesthesia. Opioids can provide obtained from a number of sources but is largely some useful sedative effects, and on occasion it dependent on the filtration rate (for haemo- maybe possible to manage patients, particularly filtration devices), or the dialysis fluid rate (for the elderly, using an opioid alone. In the major- a haemodialysis device). In practice, the ity of cases, however, the doses of opioid Drug therapies specific to critical care areas 185 required to maintain adequate sedation would popular. A more recent innovation, daily seda- be excessive and result in adverse effects and tion hold, has been more enthusiastically accumulation. As a consequence, sedative received. In this method, sedation is stopped anaesthetic agents are co-prescribed to manage each morning and only reinstated once the agitation and to facilitate the ventilation patient recovers, and if there is an ongoing clin- process. Finally in the rarer more serious cases it ical need. Because of its simplicity this system may also be necessary to use muscle relaxants to is very efficient at preventing oversedation, paralyse the patient if they fail to synchronise however it cannot be used in patients whose with the ventilator, or if paralysis is required to clinical management is likely to be comprom- control symptoms. ised if sedation is removed (e.g. head injuries, Because most of the drugs used undergo renal high ventilation requirements). In these cases excretion or hepatic metabolism, oversedation the adjustment of dosing reverts to traditional due to accumulation of the parent drug and/or methods (e.g. sedation scoring), observing active metabolites is a risk in ICU patients as physiological markers (e.g. raised heart rate/ kidney or liver impairment is relatively com- blood pressure), and the experience of the monplace. To some extent this accumulation is clinical team. unavoidable (e.g. where a patient’s clinical con- dition requires high dosages or if a prolonged Opioids period of sedation is necessary), but over- sedation can be minimised by a number of Strong opioids are the foundation for any seda- strategies, such as use of drugs that are less likely tion and analgesia used in ICU. The agents in to accumulate, regular review of dosages, seda- most common use are morphine, fentanyl, tion scoring and daily sedation holds. alfentanil and, more recently, remifentanil. The characteristics of individual sedative Morphine is the most popularly used opioid agents will be discussed below but as a general- in ICUs in the UK because it is inexpensive and ity those that are the least prone to accumula- provides excellent analgesia in patents with tion (e.g. alfentanil, remifentanil, propofol) are good renal and liver function. The parent drug also the most expensive. This means that finan- undergoes hepatic metabolism to a series of cial considerations inevitably come into play pharmacologically very active metabolites: when deciding the optimal regimen for seda- morphine-3-glucuronide (M3G) and morphine- tion, as these drugs form the major component 6-glucuronide (M6G). These metabolites are of many ICUs’ expenditure. On a more practical excreted by the kidney, and accumulate in renal level, the majority of ICUs prefer to use a lim- impairment, but are both significantly cleared ited range of agents to manage patients as this during haemofiltration or haemodialysis. reduces the likelihood of staff being unfamiliar Because of this profile, morphine needs careful with the drugs in use and consequently being prescribing and administration in patients more likely to use them inefficiently. As a result, with renal impairment to prevent significant in practice the choice of drug is affected by fac- accumulation. On occasion this can delay awak- tors other than duration of action and tendency ening for many days. Regular dosage review, to accumulate. daily sedation holds and, where appropriate, renal replacement therapy are essential to pre- vent significant accumulation. Strategies to reduce oversedation Fentanyl duration of effect is affected as Sedation scoring is a numerical system of much by the duration of use as by any other fac- recording how sedated a patient is. There are tors. In the early phase of treatment the dura- many scoring systems in use, but they are only tion is short because the agent redistributes useful if clinical staff use the data to tailor extensively throughout the body and the drug dosages on a regular basis. On many occasions itself is metabolised by hepatic metabolism this does not happen as well as it should, and (half-life ~2–12 hours). Used for short periods, consequently sedation scoring is not universally fentanyl does not accumulate clinically, but if 186 Chapter 14 • Renal pharmacy in critical care longer term infusions are used, redistribution strated to be responsible for significantly from central compartments and its slow excre- delayed awakening. As a result, this agent tion may result in delayed recovery. This effect should be used with caution in patients with is seen in all patients and is not significantly renal impairment, particularly for long dura- affected by renal function. tions of sedation. Alfentanil has a short duration of action, a Lorazepam has a longer duration of action small volume of distribution, is hepatically than midazolam but its metabolites are pharma- metabolised to inactive metabolites, but is cologically inactive. Because of its long half-life expensive compared with morphine and fen- there is a significant propensity for accumulation tanyl. Recovery from alfentanil is unlikely to be in any patient, but this is unlikely to be aggra- delayed due to renal impairment, but can be vated by renal impairment. As a result, lorazepam delayed in hepatic impairment. It is probably is increasingly been seen as an alternative sed- the agent of choice in renal impairment but ative agent to midazolam. However there have many units choose to use other agents for been concerns about adverse effects secondary to economic reasons. its excipient agent propylene glycol causing Remifentanil is metabolised by serum hyperlactaemia and other metabolic effects. esterases, and does not accumulate in renal or Propofol is the shortest acting anaesthetic liver impairment. It has a very short duration of available and it provides an excellent quality of action but is expensive compared with all of the sedation that is rapidly reversible in the major- other opioids. To date there is limited experi- ity of patients. It rarely accumulates in patients, ence with the use of this agent, and it is prin- including those with renal impairment, because cipally being used because of the quality of its it undergoes metabolism, predominantly by the sedation in difficult to manage patients rather liver to inactive substances. Until recently than its duration of action and the reduced risk propofol was an expensive agent and its use was of accumulation, which is most likely to be limited because of cost, however since the most advantageous in patients with significant expiry of its patent this agent is now relatively liver dysfunction. cheap and its use is increasing. It is probably the The pharmacokinetics of the main medica- agent of choice in patients with renal dysfunc- tions used in the ICU are summarised in tion. The major concerns with propofol are its Table 14.1. hypotensive effect and the so called ‘propofol infusion syndrome’ that has been reported in patients receiving very high doses over a pro- Sedative anaesthetic agents longed period of time. In the UK the commonest sedative agents used are the benzodiazepines (midazolam, and less Muscle relaxants commonly lorazepam) and propofol. A range of other sedatives may also be used in specific A range of non-depolarising muscle relaxants cases or where sedation is difficult to achieve. are available for use in the ICU, most units Discussion of these agents (ketamine, halo- using atracurium, vecuronium, or less com- peridol, clonidine, isoflurane) is beyond the monly cisatracurium and rocuronium. scope of this text. Atracurium is a short-acting agent with a Midazolam is the most widely used sedative duration of action of 20 minutes. It undergoes agent in the UK. When used for short-term systemic metabolism by Hofmann elimination sedation and in patients with good renal and to largely inactive metabolites, and it does not liver function it provides excellent quality accumulate in renal or liver dysfunction. One of sedation and is not associated with delayed its metabolites, laudanosine, does accumulate awakening. Midazolam undergoes hepatic markedly in renal failure but there is little evid- metabolism to a series of metabolites, princip- ence that this causes any clinically relevant side- ally 1-hydroxymidazolam glucuronide. This has effects. weak sedative effects but accumulates signific- Vecuronium has a slightly longer duration of antly in renal impairment and has been demon- action than atracurium. It is metabolised in the Drug therapies specific to critical care areas 187

Table 14.1 Pharmacokinetics of intensive care unit medications

Drug Pharmacokinetics Duration of action Effect of renal failure

Alfentanil Hepatic, inactive Half-life 1–4 hours Minimal. Small increased in free metabolites fraction, therefore doses slightly reduced Fentanyl Redistribution, hepatic Half-life 2 hours. Duration Minimal metabolism increases with time due to redistribution Morphine Hepatic, active metabolites Half-life morphine is Significant risk of accumulation of (morphine glucuronides) 1–4 hours active metabolites excreted renally Half-life morphine glucuronides is 4–50 hours Remifentanil Systemic metabolism by 3–10 minutes. Minimal serum esterases Metabolites essentially inactive Lorazepam Hepatic metabolism, Half-life 5–10 hours Small increase in duration glucuronide metabolite is inactive Midazolam Hepatic metabolism, active 1.5–12 hours Significant risk of accumulation of metabolite (1-OH midazolam) active metabolite Propofol Predominantly hepatic 30–60 min Minimal metabolism Atracurium Systemic metabolism – 20–30 min Minimal Hofmann elimination Metabolite laudanosine not muscle relaxant Vecuronium Hepatic metabolism to Half-life 0.5–1.2 hours Accumulation of active metabolites active metabolites

liver to a series of metabolites, including 3- ber of strategies are used to minimise this risk, hydroxy-vecuronium, which has approximately including prophylactic H2-antagonists, proton 50% the activity of the parent drug. These pump inhibitors, sucralfate, and early establish- metabolites are excreted renally and can cause ment of enteral feeding. delayed recovery in patients with kidney The best evidence is for the use of H2- impairment. As a result, vecuronium is probably antagonists or sucralfate. Both treatments not the first choice muscle relaxant in renal fail- reduce the incidence of stress ulceration to ure, but it maybe used if patients start to similar degrees, but there is a tendency towards develop resistance to atracurium, a phenom- higher rates of hospital-acquired pneumonia in enon that is fairly common. patients receiving H2-antagonists. Despite this evidence most ICUs use intravenous H2-blockers because they are easier to administer than Prevention of stress ulceration enteral sucralfate which can cause complica- tions with prolonged use in conjunction with ICU patients, particularly those receiving enteral feeding. Also, because it contains mechanical ventilation, have an increased risk aluminium there have been concerns expressed of developing gastric and duodenal ulcers due to about the use of sucralfate in patients with the physiological stress of their illness. A num- ARF because of the known association of 188 Chapter 14 • Renal pharmacy in critical care aluminium-containing medicines with renal depending on dosage (e.g. epinephrine (adrena- bone disease and dialysis dementia in patients line), dopamine). Readers should refer to other with ESRD. The H2-antagonist most commonly sources for a more detailed insight into the clin- used is ranitidine 50 mg three times daily ical use of these agents, but as most of these (reduced to twice daily in severe renal impair- agents have extremely short half-lives and are ment). Sucralfate is given enterally at a dose of metabolised systemically, general renal impair- 1 g every 4–6 hours. ment has little effect on their duration of Of the other treatments, enteral feeding is action. The only exception is the phosphodi- widely used, and is probably effective once the esterase inhibitors milrinone and enoximone patient has started to absorb food, as this that have a longer duration of action and do indicates a working and adequately perfused accumulate in renal failure. These agents should gastrointestinal tract. There is, however, surpris- therefore be used at lower than average doses. ingly little published evidence on its use. Proton However, as dosing is generally titrated to effect, pump inhibitors are increasingly being used, but clinically significant accumulation rarely occurs. with little evidence, and until proven otherwise there will remain concerns about the incidence of pneumonia as this is probably related to General comments on administering changes in gastric pH. drugs to patients with renal impairment Thromboprophylaxis The rules for prescribing drugs in renal impair- Intensive care unit patients typically have a ment are the same as you would apply in any higher risk of thromboembolic complications other clinical area, but a few specific points of because of their pre-existing or presenting con- guidance are worth considering. ditions (e.g. pneumonia, cardiac disease). As a A large number of drugs are administered by result, most units will give the higher doses of intravenous injection, and this can impose a thromboprophylaxis (i.e. enoxaparin 40 mg significant fluid input particularly in sicker daily, dalteparin 4000 units daily) to all patients patients receiving multiple infusions. It may with ‘normal’ clotting function. Patients with therefore be necessary to try to restrict fluid ARF may, however, require either a lower dose intake. A number of options should be consid- of prophylactic agent because of the increased ered. First you should attempt to give medicines half-life of some heparin products in renal by enteral routes if this is feasible, and does risk impairment (e.g. enoxaparin). Alternatively, impaired absorption for medicines that are crit- treatment may be omitted because they are ical for recovery. If you need to fluid restrict you receiving concurrent anticoagulant medication should consult the SPC and where necessary as part of their renal replacement therapy. minimum infusion guides (e.g. UKCPA Critical Care Group Minimum infusion volumes for fluid restricted critically ill patients4). This latter ‘Inotropic’ therapy document gives largely anecdotal advice but is invaluable. A wide range of agents are used to optimise Finally, when managing patients who are cardiovascular function in critically ill patients. extremely sick and have immediately life-threat- These range from vasopressor agents such as ening infections etc., if dosage reductions are norepinephrine (noradrenaline), phenylephrine necessary, it is worth delaying these to ensure and argipressin that are used to increase vas- that the patient receives sufficient dose of a cular tone and blood pressure, through drug to give them a chance of responding. In inotropic/vasodilator agents such as dobuta- practice, most pharmacists will delay dosage mine, enoximone, milrinone that are used in reductions until the following day as this period cardiogenic shock, to mixed agents that have of ‘overdosing’ is highly unlikely to be harmful intropic, vasodilating and vasopressor effects to the patient. References 189 C CASE STUDY

MF is a 19-year-old man weighing 70 kg who was admitted to the ICU with severe sepsis second- ary to presumed meningococcal meninigitis. On admission he is severely hypotensive, and anuric. He is prescribed cefotaxime and benzylpenicillin, is sedated for ventilation, and is commenced on norepinephrine (noradrenaline) to increase his blood pressure. His blood results on admission give a creatinine of 123 µmol/L and a urea of 7.3 mmol/L.

Q1. Assess his renal function, and decide whether it will be necessary to adjust the dosage of his antibiotics and any other medications.

Q2. The standard protocol for sedating patients on the ICU uses a combination of morphine and midazolam. The clinical staff are concerned about drug accumulation with these agents. What are the options to prevent accumulation? Twelve hours following admission, MF remains anuric, has a significant metabolic acidosis (pH 7.21, lactate 5.3 mmol/L), requires norepinephrine (noradrenaline) to maintain adequate blood pressure, and ventilatory support. Continuous veno-venous haemofiltration is commenced.

Q3. The CVVHF system is configured to an ultrafiltration rate of 30 mL/kg/h. Are any dosage modifications required for the drugs mentioned above and for the following additional agents – enoxaparin SC 40 mg once daily, ranitidine IV 50 mg three times daily, and IV insulin infusion to maintain a blood glucose of 5–8 mmol/L? The consultant prescribes drotrecogin alfa (activated protein C) as treatment for severe sepsis, at a dose of 24 µg/kg/h.

Q4. Are any dosage modifications required for this agent, and are any additional precautions necessary?

Q5. What options are available to manage MF’s metabolic acidosis?

References 3. Oudemans-van-Straaten HM, Wester JPJ, dePont ACJM, Schetz MRC. Anticoagulation strategies in continuous renal replacement therapy: can the 1. Wright SE, Bodenham A, Short AIK, Turney JH. The choice be evidence based? Intensive Care Med 2006; provision and practice of renal replacement ther- 32: 188–202. apy on adult intensive care units in the United 4. UK Clinical Pharmacy Association, Critical Care Kingdom. Anaesthesia 2003; 58: 1063–1069. Group. Minimum infusion volumes for fluid 2. Pruchnicki MC, Dasta JF. Acute renal failure in restricted critically ill patients, 3rd edn. May 2006. hospitalised patients: Part II. Ann Pharmacother Available from www.ukcpa.org.uk 2002(July/August); 36: 1261–1267.

15

Pain control in renal impairment

Stephen Ashmore

The clinical efficacy of most analgesic drugs is The analgesic ladder altered by impaired renal function, not simply because of altered clearance of the parent drug, but also through accumulation of toxic or ther- Analgesic prescribing for both acute and chronic apeutically active metabolites. Some analgesic pain should always follow a stepwise approach, agents can aggravate pre-existing renal disease, with ongoing monitoring of pain severity and causing direct damage and thus altering their adjustment of doses and agents. In the acute set- excretion. ting it is important to monitor the patient regu- This chapter will cover the basic rules for pre- larly for signs of toxicity or lack of efficacy. This scribing analgesics in patient with varying is particularly important in cases of acute renal degrees of renal impairment. Published evidence failure, where renal function can change rap- of efficacy is quoted for a wide range of agents idly, resulting in increased risk of side-effects or which have been employed to treat different underdosing. types of pain. Specific renal doses for individual The first step on the analgesic ladder is to use agents can be found in The Renal Drug non-opioids such as paracetamol and non- Handbook,1 or in the relevant summary of prod- steroidal anti-inflammatory drugs (NSAIDs) (if uct characteristics (SPC). appropriate for the renal patient); if pain persists mild opioids can be added or substituted. Finally, in severe pain a strong opioid should be used, in Assessing pain combination with an NSAID if necessary.

The assessment and ongoing measurement of Analgesics and renal impairment pain are essential to diagnosing and treating painful conditions. A visual analogue scale from 0 to 10 is often used to assess pain in adults. Analgesics that exhibit the safest pharmacologi- Patients are asked to mark on a 10-cm scale the cal profile in patients with renal impairment are point which best represents their pain, where 0 alfentanil, buprenorphine, fentanyl, ketamine represents no pain and 10 the worst pain ima- and paracetamol. None of these drugs deliver a ginable (scores above 7 cm are considered to high active metabolite load or have a signifi- represent severe pain). Pictorial representations cantly prolonged clearance. of pain (smiling/sad faces) are often used for Oxycodone can usually be used without any children. Advice on recording pain scores dose adjustment in patients with renal impair- should be obtained from acute pain specialists. ment as the main metabolite, oxymorphone, is A description of the type of pain is also only weakly active, and contributes minimally important to aid prescribing. Neuropathic pain to any clinical effect. However, as hepatic blood requires a different treatment approach to other flow can be altered in uraemia the half-life of types of pain, and will be described as burning, oxycodone can be increased as hepatic clearance stabbing or shooting in nature. is reduced. The manufacturer of oxycodone

191 192 Chapter 15 • Pain control in renal impairment

Table 15.1 Recommendations for drug dosing in renal impairment

Drug Comments Recommendations References

NSAIDs and COX-2 Can affect renal function Use with caution in mild renal 4 inhibitors impairment. Avoid in severe renal impairment Gabapentin Half-life increased and plasma Dose adjustment recommended on 5 levels raised in renal impairment basis of creatinine clearance Tricyclic Amitriptyline metabolised to active Limited data; metabolite 6 antidepressants agent in liver accumulation may increase risk of side-effects Ketamine Metabolite level increased, but has Limited data; probably no dose 7 little activity adjustment required Local anaesthetics Increase in free fraction may result Risk of toxicity may be affected by 8–10 from alterations in protein binding acid–base disturbances or serum potassium levels

Adapted from ref. 2. therefore recommends cautious use of the drug The mechanism of action of paracetamol in chronic renal and hepatic disease, with a low remains unclear. It has no known endogenous starting dose.2 binding sites, and apparently does not inhibit Analgesics that have been used in patients peripheral cyclo-oxygenase activity. There is with renal impairment, but may require a reduc- increasing evidence of a central antinociceptive tion in dose include amitriptyline, clonidine, effect. Potential mechanisms for this include codeine, gabapentin, hydromorphone, lido- inhibition of a COX-2 in the CNS, or inhibition caine, methadone, morphine and tramadol. of a central cyclo-oxygenase ‘COX-3’ that is NSAIDs, dextropropoxyphene (found in co- selectively susceptible to paracetamol, and proxamol) and pethidine are not recommended modulation of inhibitory descending seroton- in patients with significant renal impairment.3 ergic pathways. Paracetamol has also been Table 15.1 summarises some recent published shown to prevent prostaglandin production, recommendations for various analgesics in independent of cyclo-oxygenase activity.11 patients with renal impairment. Practice in indi- Paracetamol is an effective adjunct to opioid vidual renal units may differ from these analgesia, opioid requirements being reduced by recommendations. 20–30% when combined with a regular regimen of oral or rectal paracetamol. The combination of paracetamol 1000 mg plus codeine 60 mg has a number-needed-to-treat (NNT) of 2.2. The Individual agents addition of an NSAID to paracetamol further improves efficacy.12 Intravenous (IV) paracetamol is an effective Paracetamol analgesic after surgery, is as effective as ketoro- lac, and is equivalent to morphine and better Paracetamol is an effective analgesic and tolerated after dental surgery, although there is antipyretic. It is absorbed rapidly and well from evidence of a ceiling effect.3 the small intestine after oral administration and Paracetamol has fewer side-effects than can be given rectally and intravenously. NSAIDs and can be used when the latter are Individual agents 193 contraindicated (e.g. patients with a history of Side-effects asthma, renal impairment or peptic ulcers). It should be used with caution or in reduced NSAID side-effects are more common with long- doses in patients with active liver disease, term use. In general, the risk and severity of alcohol-related liver disease and glucose-6- NSAID-associated side-effects is increased in the phosphate dehydrogenase deficiency. In these elderly. situations, as well as in overdose, the rate of reactive metabolite production can result in Renal function liver damage, occasionally with acute renal Renal prostaglandins regulate tubular electrolyte tubular necrosis. handling, modulate the actions of renal hor- In cases of severe renal impairment, the elim- mones, and maintain renal blood flow and ination of paracetamol is slightly delayed and glomerular filtration rate in the presence of metabolites may accumulate. It has been sug- circulating vasoconstrictors. In some clinical gested that the maximum daily dose of para- conditions, including hypovolaemia and dehyd- cetamol for chronic use in patients with end ration, high circulating concentrations of vaso- stage renal failure (ESRF) should be reduced to constrictors increase production of intrarenal 3g.13 However, in practice, few renal units vasodilators including prostacyclin – mainten- adopt this strategy. ance of renal function may then depend on prostaglandin synthesis and thus can be sensit- ive to brief NSAID administration. Non-steroidal anti-inflammatory drugs Diclofenac has been shown to affect renal function in the immediate post-operative period NSAIDs have analgesic, anti-inflammatory and after major surgery and administration of other antipyretic effects and are effective analgesics in potential nephrotoxins, such as gentamicin, can a variety of acute pain states. Unfortunately, increase the renal effects of ketorolac. significant contraindications and adverse effects The risk of adverse renal effects of NSAIDs limit the use of NSAIDs in many patients, and COX-2 inhibitors is increased in the pres- including those with renal impairment. ence of factors such as pre-existing renal Prostaglandins have many physiological impairment, hypovolaemia, hypotension, use of functions including gastric mucosal protection, other nephrotoxic agents and angiotensin- 3 and renal tubular function which are mainly converting enzyme (ACE) inhibitors. regulated by COX-1 and are the basis for many of the adverse effects associated with NSAID use. Platelet function Tissue damage induces COX-2 production, lead- NSAIDs inhibit platelet function by their effect ing to synthesis of prostaglandins that result in on COX-1. In patients with bleeding problems pain and inflammation. COX-2 may be ‘consti- or receiving anticoagulants, there is an tutive’ in some tissues, including the kidney. increased risk of significant surgical blood loss NSAIDs inhibit both COX-1 and COX-2. Aspirin after NSAID administration. See also Cardio- acetylates and inhibits cyclo-oxygenase irre- vascular risks of NSAIDs and COX-2 inhibitors versibly but NSAIDs are reversible inhibitors of below. the enzymes. The COX-2 inhibitors have been developed to inhibit selectively the inducible Gastrointestinal side-effects form. Acute gastroduodenal damage and bleeding can When given in combination with opioids occur with short-term NSAID use. The risk is after surgery, NSAIDs result in better analgesia increased with higher doses, a history of peptic and reduce opioid consumption. The addition ulceration, use for more than 5 days and in the of an oral NSAID to paracetamol also improves elderly. After 5 days of naproxen and ketorolac analgesia.3 use in healthy elderly subjects, ulcers were found on gastroscopy in 20% and 31% of cases respectively.14 194 Chapter 15 • Pain control in renal impairment

The gastric and duodenal epithelia have vari- NSAIDs.18 The lack of antiplatelet effects may be ous protective mechanisms against acid and an advantage for the patient with bleeding enzyme attack and many of these involve problems, or when anticoagulants are given. prostaglandin production. Chronic NSAID use is Significant research has been undertaken into associated with peptic ulceration and bleeding whether COX-2 inhibitors can produce a tend- and the latter may be exacerbated by the ency to thrombosis because they inhibit antiplatelet effect. It has been estimated that endothelial prostacyclin production but spare the relative risk of perforations, ulcers and platelet thromboxane synthesis and aggregation. bleeds associated with NSAIDs is 2.7 compared The VIGOR study, in which patients on low- with people not consuming NSAIDs.15 dose aspirin were excluded, found an increased risk of myocardial infarction for patients given Bronchospasm rofecoxib compared with naproxen.19 Rofecoxib Aspirin-exacerbated respiratory disease (AERD) was withdrawn from clinical practice in 2004 is a recognised problem in individuals with because of further concerns about the risks of asthma, chronic rhinitis and nasal polyps. AERD cardiovascular events including myocardial affects 10–15% of people with asthma, can be infarction and stroke.20 See also Cardiovascular severe and there is a cross-sensitivity with risks of NSAIDs and COX-2 inhibitors below. NSAIDs but not selective COX-2 inhibitors.16 A history of AERD is a contraindication to NSAID Gastrointestinal side-effects use, although there is no reason to avoid Large outcome studies have demonstrated that NSAIDs in other people with asthma. COX-2 inhibitors produce less clinically signi- ficant peptic ulceration than NSAIDs. Both rofecoxib and celecoxib have been associated with a substantial reduction in endoscopic Cyclo-oxygenase-2 selective inhibitors ulcers compared with NSAID comparators.3 In (COX-2 inhibitors) the VIGOR study all upper gastroinstestinal events were reduced with rofecoxib compared These agents selectively inhibit the inducible with naproxen. cyclo-oxygenase enzyme COX-2, and spare COX-1. They offer the potential for effective Bronchospasm analgesia with fewer side-effects than NSAIDs. Studies in patients with AERD have shown that At the time of writing there are two COX-2 analgesic doses of COX-2 inhibitors do not pro- inhibitors, or coxibs, on the UK market – cele- duce bronchospasm in these patients.16 coxib and etoricoxib.

Side-effects Cardiovascular risks of COX-2 inhibitors

Renal function Following the results from studies such as COX-2 is continuously produced in the kidney VIGOR and the Adenoma Prevention with and is increased in response to alterations in Celecoxib (APC) Trial21 the Medicines and intravascular volume. COX-2 has been impli- Healthcare Products Regulatory Agency (MHRA) cated in maintenance of renal blood flow, medi- reported on the safety of COX-2 inhibitors in ation of renin release and regulation of sodium February 2005. The conclusions are set out excretion. COX-2 inhibitors and NSAIDs have below: similar adverse effects on renal function.17 The evidence suggests that selective COX-2 inhibitors, as a class, may cause an increased Platelet function risk of thrombotic events (e.g. myocardial Platelets produce only COX-1, and hence COX- infarction and stroke) compared with placebo 2 selective inhibitors do not impair platelet and some NSAIDs, and the risk may increase function. The use of COX-2 inhibitors reduces with dose and duration of exposure. It is not surgical blood loss in comparison with possible to quantify the risk precisely, but it is Individual agents 195 considered unlikely to exceed one extra serious particularly naproxen preferentially inhibit thrombotic event per 100 patient years, over the COX-1. rate for no treatment. COX-1 must be inhibited by at least 95% It was advised that: before platelet function is affected in vivo. Therefore only drugs that are highly selective • Patients with established ischaemic heart dis- for COX-1, such as naproxen are likely to be ease or cerebrovascular disease should be cardioprotective. Whether COX-2 ‘preferential’ switched to alternative treatment: in addi- drugs such as etodolac and meloxicam share tion, the existing contraindication for severe the same pro-thrombotic effects as the highly heart failure has been extended to include selective ‘coxibs’ is uncertain, but from the per- moderate heart failure (New York Heart spective of platelet aggregation there appears Association (NYHA) class II–IV). little evidence to suggest that they would differ. • For all patients the balance of gastrointestinal The association between exposure to NSAIDs and cardiovascular risk should be considered and coxibs and subsequent risk of myocardial before prescribing a COX-2 inhibitor, particu- infarction has been investigated in a large num- larly for those with risk factors for heart dis- ber of epidemiological studies. The tentative ease and those taking low-dose aspirin, for observation from the epidemiological data whom gastrointestinal benefit has not been alone is that: clearly demonstrated. • The lowest effective dose of COX-2 inhibitor • In relation to non- or ‘remote’ use of NSAIDs, should be used for the shortest necessary naproxen usage was associated with a slightly period. Periodic re-evaluation is recom- protective or null effect with regard to mended, especially for osteoarthritis patients myocardial infarction. who may only require intermittent treatment. • In relation to non- or ‘remote’ use of NSAIDs, • Gastroprotective agents should be considered ibuprofen usage was associated with a for patients switched to non-selective slightly increased or null effect with regard to NSAIDs.22 myocardial infarction. • In relation to non- or ‘remote’ use of NSAIDs, A systematic review and meta-analysis of ran- diclofenac usage was associated with an domised double-blind trials of celecoxib has elevated risk with regard to myocardial shown an increased risk of myocardial infarc- infarction. tion with celecoxib therapy, which confirms the advice given above.23 The evidence of an increased thrombotic risk Considering the high prevalence of cardio- associated with NSAIDs is much less clear than vascular disease in patients with established for coxibs. Based on the available evidence, any renal disease it would seem appropriate to avoid increased risk is likely to be small and associated the use of COX-2 inhibitors in such patients. with continuous longer term treatment and high doses. Most of the available data relate to naproxen, Cardiovascular risks of non-steroidal anti- ibuprofen and diclofenac. Some trials have inflammatory drugs shown naproxen to have a lower thrombotic risk than selective COX-2 inhibitors. However, In August 2005 the MHRA reported on the data on the risk for ibuprofen and diclofenac, safety of NSAIDs. The conclusions are set out relative to COX-2 inhibitors, are less clear. The below: absence of useful data for other NSAIDs should As NSAIDs exhibit a broad range of COX-2/ not be taken to imply a lower risk for these COX-1 selectivity, it would be important to products. know if differing degrees of selectivity correlate The MHRA concluded that overall, the data with differing biological effects. The three most on thrombotic risk with traditional NSAIDs are widely used standard NSAIDs have differing insufficient to warrant changes in current pre- levels of COX1/2 selectivity: diclofenac is scribing practice. Any prescribing decision COX-2 ‘preferential’, whereas ibuprofen and should be based on the overall safety profile of 196 Chapter 15 • Pain control in renal impairment

NSAIDs (particularly gastrointestinal safety pro- Despite the lack of published safety data for file), and the individual risk factors of the dihydrocodeine in patients with renal impair- patient. For all NSAIDs, the lowest effective dose ment, and the reported side-effects of codeine in should be used for the shortest period of time this population, both drugs are widely used in necessary in order to control symptoms. renal units. Codeine is particularly popular, Although platelet aggregation studies have often combined with paracetamol. Patients demonstrated an interaction between aspirin receiving regular co-codamol 30/500 should be and ibuprofen, a clinically important effect has monitored for side-effects. not been clearly demonstrated in epidemiologi- cal studies or clinical trials. There is less evid- ence for such an interaction between naproxen Dextropropoxyphene and aspirin.24 Dextropropoxyphene is a weak opioid with an NNT of 7.7. In the UK it has been used in com- bination with paracetamol (co-proxamol) but Opioids this combination improves pain relief by only 7.3% compared with paracetamol alone and Opioids remain the mainstay of systemic anal- increases the incidence of dizziness.3 gesia for the treatment of moderate to severe The major metabolite of dextropropoxy- acute pain. Interpatient opioid requirements phene is nordextropropoxyphene which is vary greatly, and opioid doses therefore need to renally excreted; accumulation of nordextro- be titrated to suit each patient. In adult patients, propoxyphene can lead to CNS, respiratory and age rather than weight is the better predictor of cardiac depression. opioid requirements (Table 15.2). 25 At the time of writing co-proxamol has been All full opioid agonists given in equi- withdrawn from the UK market. analgesic doses produce the same analgesic effect, although such equi-analgesic doses are difficult to determine due to interindividual Morphine variabilities in kinetics and dynamics.34 Most available data do not suggest that any Morphine remains the most widely used opioid one opioid is superior to another, either in for the management of pain and the standard terms of better pain relief, differences in side- against which other opioids are compared. effects or patient satisfaction, but rather that Morphine-6-glucuronide (M6G) and morphine- some opioids may be better in some patients; 3-glucuronide (M3G), the main metabolites of although pethidine has been reported to have a morphine, are formed by morphine glucuron- higher incidence of nausea and vomiting com- idation, primarily in the liver. M6G is a mu- pared with morphine.3 opioid agonist and may be more potent than morphine with morphine-like effects, including analgesia. M3G has very low affinity for opioid Codeine and dihydrocodeine receptors, has no analgesic activity and animal Codeine is classified as a weak opioid but the studies have shown that it may antagonise the molecule itself is devoid of analgesic activity. analgesic effects of morphine and be responsible The main metabolite of codeine is codeine-6- for neurotoxic symptoms sometimes associated glucuronide, which has a similar potency to the with high doses of morphine. parent drug and is renally excreted; 2–10% of a Both M6G and M3G are dependent on the dose is metabolised to morphine, which kidney for excretion. Impaired renal function, accounts for most of the analgesic effect of the oral route of administration (first-pass codeine. metabolism), higher doses and increased patient Dihydrocodeine is a semi-synthetic derivat- age are predictors of higher M3G and M6G ive of codeine, with an analgesic effect inde- concentrations.35 pendent of its metabolism to dihydromorphine. Individual agents 197

Table 15.2 Recommendations for opioid dosing in renal impairment

Drug Comments Recommendations References

Alfentanil No active metabolites. Alterations No dose adjustment required 26, 27 in protein binding may lead to increased free drug Buprenorphine Inactive and weakly active No dose adjustment required 26, 27 metabolites. Mainly biliary excretion Codeine Prolonged sedation and respiratory Dose adjustment recommended. 26, 27 arrest reported. Neuro-excitation Use an alternative if possible with normal doses Dextropropoxyphene Accumulation of active metabolite Dose adjustment recommended. 27, 28 can lead to CNS and CVS toxicity Use an alternative if possible Dihydrocodeine Metabolism probably similar to Insufficient evidence: use not 26 codeine recommended Fentanyl No active metabolites No dose adjustment required 27 Hydromorphone Neurotoxicity from accumulation of Dose adjustment recommended. 27, 29 metabolite possible Use an alternative agent if high doses needed Methadone Mainly inactive metabolites, but Dose adjustment recommended 26, 27 20% excreted unchanged via in severe renal impairment kidneys Morphine Major metabolites excreted via Dose adjustment recommended. 26, 27, 30 kidneys. Delayed sedation and Use an alternative agent if high neurotoxicity from accumulation of doses needed metabolites

Oxycodone Oxymorphone metabolite has little No dose adjustment required 28 clinical effect Pethidine Accumulation of norpethidine can Dose adjustment required. Use 26, 27, 31, 32 lead to seizures of alternative recommended Tramadol Increased effects from active Dose adjustment recommended. 27, 33 metabolite Use an alternative agent in significant renal impairment

Adapted from ref. 2.

Diamorphine phine that are thought to be responsible for the analgesic effects of diamorphine. There is no Diamorphine (diacetylmorphine) is rapidly difference between parenteral diamorphine hydrolysed to monoacetylmorphine (MAM) and and morphine in terms of analgesia and side- morphine; diamorphine and MAM are more effects.36 lipid soluble than morphine and penetrate the CNS more rapidly, although it is MAM and mor- 198 Chapter 15 • Pain control in renal impairment

Fentanyl with mild to moderate renal impairment, and contraindicate its use in dialysis patients due Fentanyl is increasingly used in the treatment of to an increase in the elimination half-life of acute pain because of its lack of active meta- oxycodone. bolites and fast onset of action. The develop- ment of fentanyl patches for the treatment of chronic pain has increased the use of this Pethidine drug, and is a useful addition to the analgesic formulary for patients with all degrees of renal Pethidine is a synthetic opioid still widely used impairment. even though it has multiple disadvantages. Despite a common belief that it is the most effective opioid in the treatment of renal colic, Hydromorphone it is no better than morphine. Similarly, pethid- Hydromorphone is a derivative of morphine ine and morphine have similar effects on the and is between five and ten times as potent as sphincter of Oddi and biliary tract and there is morphine. There is little difference between no evidence that pethidine is better in the treat- 3 hydromorphone and other opioids in terms ment of biliary colic. of analgesic efficacy or adverse effects. The Accumulation of the active metabolite nor- main metabolite of hydromorphone is pethidine is associated with neuroexcitatory hydromorphone-3-glucuronide, a structural effects that range from nervousness to tremors, analogue of M3G and, like M3G, it is depend- twitches and seizures. As impaired renal func- ent on the kidney for excretion, has no anal- tion increases the half-life of norpethidine, gesic action and can lead to dose-dependent patients in renal failure are at increased risk of neurotoxic effects.3 norpethidine toxicity. Naloxone does not reverse and may increase the problems related to norpethidine toxicity. Methadone Methadone is commonly used for the mainten- ance treatment of patients with an addiction to Tramadol opioids because of its good oral bioavailability Tramadol is an atypical centrally acting anal- (60–95%), high potency and long duration of gesic because of its combined effects as an action. In addition, its lack of active meta- opioid agonist and a serotonin and noradrena- bolites, low cost and additional effects as an N- line reuptake inhibitor. It is listed as a weak methyl-D-aspartate (NMDA) receptor antagonist opioid by the World Health Organization. and selective serotonin reuptake inhibitor (SSRI) Tramadol is effective in the treatment of have led to its increasing use in the treatment neuropathic pain with an NNT of 3.5. Its of cancer and chronic non-cancer pain. Its use adverse effect profile is different from other in acute pain treatment is limited by its long opioids. The risk of respiratory depression is and unpredictable duration of action and the significantly lower at equianalgesic doses. risk of accumulation. Significant respiratory depression has been described in patients with severe renal failure, Oxycodone most likely due to accumulation of the meta- bolite O-desmethyltramadol (M1), which has Oxycodone is a potent opioid agonist, with higher affinity for the opioid receptor. approximately 2–3 times the potency of mor- In addition, tramadol has limited effects on phine. Oxycodone is metabolised in the liver gastrointestinal motor function and causes less primarily to noroxycodone and oxymorphone; constipation than morphine. Nausea and vom- oxymorphone is weakly active but contributes iting are the most common adverse effects and minimally to any clinical effect. Despite the occur at rates similar to other opioids. Tramadol minimal activity of oxymorphone the manufac- does not increase the incidence of seizures com- turers of oxycodone caution its use in patients pared with other analgesic agents.3 Individual agents 199

Adverse effects of opioids to the low amounts of active drug that are excreted via the kidneys, no dose reduction is Common adverse effects of opioids are sedation, required for ketamine in patients with renal pruritus, nausea, vomiting, slowing of gastro- impairment. intestinal function and urinary retention. Clinically meaningful adverse effects of opioids are dose-related; once a threshold dose is Antidepressant drugs reached, every 3–4 mg increase of morphine- Antidepressants are effective in the treatment of equivalent dose per day is associated with one a variety of chronic neuropathic pain states additional adverse event or patient-day with (Table 15.3). There is also good evidence for the such an event.37 effect of antidepressants in chronic headaches Respiratory depression can usually be with an NNT of 3.2 and for pain relief, but not avoided by careful titration of the dose against improved function, in chronic back pain.3 effect. As respiratory depression is almost always Currently the use of antidepressants for acute preceded by sedation, the best early clinical neuropathic pain is mainly based on extrapola- indicator is increasing sedation.38 tion of the above data. However, amitriptyline Those opioids that are dependent on renal and venlafaxine are effective in the treatment of elimination should be used with caution in established neuropathic pain following breast patients with renal impairment, and the poss- surgery. In addition there is a possible pre- ibility of drug-related side-effects should always ventive effect – given before and continued after be borne in mind. The idea of ‘small doses surgery, venlafaxine significantly reduced the not very often’ with titration to effect should incidence of chronic pain at six months, and form the basis of opioid dosing for many of amitriptyline given to patients with acute these drugs in a population at risk of opioid herpes zoster reduced the incidence of posther- accumulation. petic neuralgia at six months.3 Clinical experience in chronic pain suggests that tricyclic antidepressants (TCAs) should be Adjuvant drugs

N-Methyl-D-aspartate receptor antagonists Table 15.3 Antidepressants for the treatment of N-Methyl-D-aspartate (NMDA) receptors are diabetic neuropathy and postherpetic neuralgia present in peripheral and central nerves. (placebo-controlled trials) Activation of NMDA receptors is linked to learn- ing and memory, neural development and acute Efficacy NNT (95% CI) and chronic pain states. Diabetic neuropathy The NMDA receptor antagonist ketamine is TCAs 2.4 (2.0–3.0) used clinically. In chronic pain states such as SSRIs 6.7 (3.4–435) central pain, complex regional pain syndrome, Postherpetic neuralgia fibromyalgia and ischaemic and neuropathic TCAs 2.1 (1.7–3.0) pain, there is moderate to weak evidence that Minor adverse effects NNH (95% CI) Pooled diagnoses ketamine, either as the sole agent or in com- TCAs 2.8 (2.0–4.7) bination with other analgesics, improves pain SSRIs No data available and/or decreases the requirement for other anal- Major adverse effects NNH (95% CI) gesic agents.39 Pooled diagnoses In patients with severe pain that was incom- TCAs 17.0 (10–43) pletely relieved by morphine, the addition of SSRIs Not different from placebo ketamine to the morphine regimen provided rapid, effective and prolonged analgesia.40 NNT, number-needed-to-treat; NNH, number-needed-to-harm; TCA, Most of a dose appears in the urine as tricyclic antidepressant; SSRI, selective serotonin reuptake inhibitor; CI, hydroxylated and conjugated metabolites; 4% confidence interval. is excreted unchanged or as norketamine. Due Adapted from refs 41 and 42. 200 Chapter 15 • Pain control in renal impairment started at low doses, in contrast to the treatment escalation study in 19 diabetic patients. The of depression (e.g. amitriptyline 5–10 mg at most commonly reported adverse effects night). Subsequent doses can be increased were fatigue, sweating and nausea.47 slowly if needed, in order to minimise the incid- • Venlafaxine relieved the unremitting pain of ence of adverse effects. diabetic peripheral neuropathy in eight TCAs are mainly hepatically metabolised, patients who found no relief from a variety of and there is little indication for dose reduction other treatments. NSAIDs, paracetamol, car- in renal failure, although metabolite accumula- bamazepine, capsaicin and amitriptyline were tion at higher doses may increase the incidence not successful, either due to lack of efficacy or of side-effects. The cardiovascular side-effects of to intolerable side-effects. Within 2–8 days of these agents may limit their use in certain beginning treatment, all eight patients patients. responded to venlafaxine 37.5 mg twice daily Evidence for the use of SSRIs in pain, consists with dramatic relief in symptoms associated of a number of small studies and anecdotal with painful peripheral neuropathy. No seri- reports including: ous side-effects were observed.48 Eleven patients with type II diabetes mellitus and • Slight but significant improvement of dia- painful diabetic neuropathy had a 75–100% betic neuropathy symptoms was reported reduction in pain within a few days after with citalopram 40 mg daily in a small beginning venlafaxine.49 All patients had placebo-controlled study.43 been treated unsuccessfully with other med- • Duloxetine is licensed for the treatment of ications known to alleviate the pain associ- diabetic neuropathy. Duloxetine treatment ated with diabetic peripheral neuropathy. No effectively reduced neuropathic pain in adverse effects were reported. Two patients patients with diabetic neuropathy in two ran- who were pain-free stopped taking venla- domised, double-blind, placebo-controlled faxine and had a recurrence of pain 2–3 days studies.44 Duloxetine is contraindicated in later. When venlafaxine was restarted, the patients with severe renal impairment (creat- pain was relieved promptly. inine clearance <30 mL/min). The disposition of venlafaxine and its • The case of a 31-year-old woman with auto- active metabolite, o-desmethylvenlafaxine, is nomic and peripheral neuropathy secondary altered in renal disease. Venlafaxine clear- to insulin-dependent diabetes mellitus and ance is decreased by approximately 30 to major depression is reported. Fluoxetine ther- 35% in patients with renal dysfunction. Total apy was titrated up to 40 mg per day. After daily dose should be reduced by 25% in seven months without pain the patient’s pain patients with mild to moderate renal impair- and depression returned, accompanying ment. The total daily dose should be reduced deterioration in her disease state. A dosage by 50% in patients with creatinine clearance increase to 60 mg/day was quickly followed values less than 30 mL/min. The reduced by pain relief, and later followed by improve- dose could be given once a day because of ment of her depression.45 the prolonged half-life in this population.50 • A 47-year-old man with back pain and depression due to injuries to his neck Anticonvulsant drugs and back during a fall, benefited from mirt- azapine therapy.46 Mirtazapine 15 mg at bed- Anticonvulsants have been used to treat chronic time was initiated and after one month his neuropathic pain and various systematic mood improved. His back pain also decreased reviews have shown their efficacy in a variety of from a 10 to a 3 on a 10-point scale. He was neuropathic pain states (Table 15.4). able to walk greater lengths with less diffi- In acute pain after surgery, sodium valproate culty and without his legs becoming ‘weak’. is of no benefit, whereas peri-operative • 10–60 mg/day (median 40 mg/ gabapentin leads to substantial reductions in day) was effective in relieving symptoms of both post-operative analgesic requirements and diabetic neuropathy in a single-blind, dose- pain.3 References 201

Pregabalin Table 15.4 Anticonvulsants for the treatment of Pregabalin is structurally related to gabapentin diabetic neuropathy and postherpetic neuralgia and is licensed for the treatment of peripheral (placebo-controlled trials) neuropathic pain. Like gabapentin it requires significant dosage reduction in renal impair- Efficacy NNT (95% CI) ment.56 It has been shown to be an effective Diabetic neuropathy 2.7 (2.2–3.8) analgesic agent in placebo-controlled trials, but Postherpetic neuralgia 3.2 (2.4–5.0) trials comparing it with other agents are Minor adverse effects NNH (95% CI) awaited. Pooled diagnoses 2.7 (2.2–3.4) Major adverse effects NNH (95% CI) Pooled diagnoses Not different from placebo Sodium valproate Sodium valproate has an NNT of 3.5 for at least NNT, number-needed-to-treat; NNH, number-needed-to-harm; CI, a 50% reduction in migraine frequency.57 The confidence interval. NNHs for nausea, tremor, dizziness and drowsi- Adapted from ref. 42. ness were 3.3, 6.2, 6.5 and 6.3 respectively. The NNH for withdrawals due to adverse effects with sodium valproate was 9.4. In patients with renal insufficiency, it may be Carbamazepine necessary to decrease the dosage due to reduced In a systematic review, carbamazepine was albumin levels and associated increased free found to have an NNT of 2.6 in trigeminal drug levels. As monitoring of plasma concentra- neuralgia and 3.3 in diabetic neuropathy.51 The tions may be misleading, dosage should be number-needed-to-harm (NNH) was 3.4 for adjusted according to clinical monitoring.58 minor adverse effects and 24 for severe adverse effects. Membrane stabilisers Only 1% of a dose is excreted renally. Carbamazepine has been reported as causing Intravenous lidocaine infusions are effective in reversible acute renal failure, although this is reducing pain in chronic neuropathic pain rare.52 conditions.3 Overall, the strongest evidence is for use of membrane stabilisers in pain due to Gabapentin peripheral nerve trauma.59 In the same review the NNTs for gabapentin Mexiletine has been used in the treatment of ranged between 3.2 and 3.8 in the treatment of refractory neuropathic pain in patients who chronic neuropathic pain states. The NNH for a have responded to IV lidocaine, with which it minor adverse effect compared with a placebo has structural similarities. Oral mexiletine was 2.6 (2.1–3.3). Gabapentin is also effective in showed limited efficacy with an NNT of 10 for the treatment of post-amputation phantom diabetic neuropathy. It has been shown to have pain.53 It should be used with caution in normal clearance in patients with creatinine patients with renal impairment, and doses clearances >10 mL/min, with up to 15% of a reduced to avoid accumulation.54 dose excreted unchanged in the urine.60

Lamotrigine The NNT of , based on a limited number of studies in trigeminal neuralgia, is 2.1 References (1.3–6.1).51 In single-dose studies in subjects with end stage renal failure, plasma concentra- 1. Ashley C, Currie A. The Renal Drug Handbook, 2nd tions of lamotrigine were not significantly edn. Oxford: Radcliffe Medical Press, 2004. altered. However, accumulation of the glu- 2. Oxycontin summary of product characteristics. curonide metabolite is to be expected; caution Napp Pharmaceuticals Ltd, 2005. should therefore be exercised in treating 3. Australian and New Zealand College of patients with renal failure.55 Anaesthetists and Faculty of Pain Medicine. Acute 202 Chapter 15 • Pain control in renal impairment

C CASE STUDY

A 33-year-old female haemodialysis patient has a 2-year history of left hand pain following a fractured metacarpel after falling off a horse. The pain worsens in cold weather. Her hand is stiff on movement which causes pain and restricts her activities. Her hand swells intermittently and becomes blue and cold. She has been told that the pain has no physical cause and that she must be imagining it. She feels that no one believes her and is concerned she may be going mad.

Previous medical history: • IgA nephropathy • Low mood • Gastric ulcer three months ago.

Drug history: • Fluoxetine 20 mg once daily • Bisoprolol 2.5 mg once daily • Calcichew 2 three times daily • Alfacalcidol 0.25 µg once daily • Vitamin B Co Forte 2 once daily • Lansoprazole 30 mg once daily.

Presenting complaint: Seems drowsy, withdrawn and low in mood. Says she sleeps very poorly. Diagnosis of complex regional pain syndrome.

Doctor prescribes: • Amitriptyline 50 mg every night • Tramadol MR 100 mg twice daily • Guanethidine blocks.

Q1. Comment on this prescription. What interventions might you make?

Pain Management: Scientific Evidence. Australian chronic renal failure. Clin Pharmacol Ther 1985; and New Zealand College of Anaesthetists, 2005. 37: 301–307. 4. Royal College of Anaesthetists. Guidelines for the 7. Koppel C, Arndt I, Ibe K. Effects of enzyme induc- Use of Non-steroidal Anti-inflammatory Drugs in tion, renal and cardiac function on ketamine the Perioperative Period. Oxford: Royal College of plasma kinetics in patients with ketamine long- Anaesthetists, 1999. term analgesosedation. Eur J Drug Metab 5. Blum RA, Comstock TJ, Sica DA et al. Pharmacokinet 1990; 15: 259–263. Pharmacokinetics of gabapentin in subjects with 8. Crews JC, Weller RS, Moss J et al. various degrees of renal function. Clin Pharmacol Levobupivacaine for axillary brachial plexus Ther 1994; 56: 154–159. block: a pharmacokinetic and clinical compari- 6. Liebermann JA, Cooper TB, Suckow RF et al. son in patients with normal renal function or Tricyclic antidepressant and metabolite levels in renal disease. Anesth Analg 2002; 95: 219–223. References 203

9. Rice AS, Pither CE, Tucker GT. Plasma concentra- adenoma prevention. N Engl J Med 2005; 352: tions of bupivacaine after supraclavicular brachial 1071–1080. plexus blockade in patients with chronic renal 22. Medicines and Healthcare Products Regulatory failure. Anaesthesia 1991; 46: 354–357. Agency. Updated advice on the safety of selective 10. McEllistrem RF, Schell J, O’Malley K et al. COX-2 inhibitors. February 2005. Interscalene brachial plexus blockade with lido- 23. Caldwell B, Aldington S, Weatherall M et al. Risk caine in chronic renal failure – a pharmacokinetic of cardiovascular events and celecoxib: a system- study. Can J Anaesth 1989; 36: 59–63. atic review and meta-analysis. J R Soc Med 2006; 11. Mancini F, Landolfi C, Muzio M et al. 99: 132–240. Acetaminophen down-regulates interleukin-1 24. Medicines and Healthcare Products Regulatory beta-induced nuclear factor-kappa B nuclear Agency. Cardiovascular safety of non-steroidal translocation in a human astrocytic cell line. anti-inflammatory drugs. Overview of key data. Neurosci Lett 2003; 353: 79–82. August 2005. 12. Rømsing J, Møiniche S, Dahl JB. Rectal and 25. Macintyre PE, Jarvis DA. Age is the best predictor parenteral paracetamol, and paracetamol in com- of postoperative morphine requirements. Pain bination with NSAIDs, for postoperative anal- 1996; 64: 357–364. gesia. Br J Anaesth 2002; 88: 215–226. 26. Davies G, Kingswood C, Street M. 13. Bennett WM, Aronoff GR, Golper TA et al. Drug Pharmacokinetics of opioids in renal dysfunc- Prescribing in Renal Failure, Philadelphia, PA: tion. Clin Pharmacokinet 1996; 31: 410–422. American College of Physicians, 1994. 27. Mercadante S, Arcuri E. Opioids and renal func- 14. Goldstein JL, Kivitz AJ, Verburg KM et al. A com- tion. J Pain 2004; 5: 2–19. parison of the upper gastrointestinal mucosal 28. AMH. Australian Medicines Handbook. Adelaide: effects of valdecoxib, naproxen and placebo in Australian Medicines Handbook Pty Ltd, 2004. healthy elderly subjects. Aliment Pharmacol Ther 29. Babul N, Darke AC, Hagen N. Hydromorphone 2003; 18: 125–132. metabolite accumulation in renal failure. J Pain 15. Ofman JJ, MacLean CH, Straus WL et al. A meta- Sympt Manage 1995; 3: 184–186. analysis of severe upper gastrointestinal compli- 30. Angst MS, Bührer M, Lötsch J. Insidious intoxi- cations of nonsteroidal antiinflammatory drugs. cation after morphine treatment in renal failure: J Rheumatol 2002; 29: 804–812. delayed onset of morphine-6-glucuronide action. 16. West PM, Fernandez C. Safety of COX-2 Anesthesiology 2000; 92: 1473–1476. inhibitors in asthma patients with aspirin hyper- 31. Stone PA, Macintyre PE, Jarvis DA. Norpethidine sensitivity. Ann Pharmacother 2003; 37: 1497– toxicity and patient controlled analgesia. Br J 1501. Anaesth 1993; 71: 738–740. 17. Curtis SP, Ng J, Yu QF et al. Renal effects of etoricoxib and comparator nonsteroidal anti- 32. Simopoulos TT, Smith HS, Peeters-Asdourian C et inflammatory drugs in controlled clinical trials. al. Use of meperidine in patient-controlled anal- Clin Ther 2004; 26: 70–83. gesia and the development of a normeperidine 18. Hegi TR, Bombeli T, Seifert B et al. Effect of toxic reaction. Arch Surg 2002; 137: 84–88. rofecoxib on platelet aggregation and blood 33. MIMS. MIMS Annual. MediMedia Australia Pty loss in gynaecological and breast surgery com- Ltd, 2004. pared with diclofenac. Br J Anaesth 2004; 92: 34. Gammaitoni AR, Fine P, Alvarez N et al. Clinical 523–531. application of opioid equianalgesic data. Clin J 19. Bombardier C, Laine L, Reicin A et al. Pain 2003; 19: 286–297. Comparison of upper gastrointestinal toxicity of 35. Klepstad P, Dale O, Kaasa S et al. Influences on rofecoxib and naproxen in patients with rheuma- serum concentrations of morphine, M6G and toid arthritis. VIGOR Study Group. N Engl J Med M3G during routine clinical drug monitoring: a 2000; 343: 1520–1528. prospective survey in 300 adult cancer patients. 20. US Food and Drug Administration (FDA). FDA Acta Anaesthesiol Scand 2003; 47: 725–731. Public Health Advisory: Safety of Vioxx. US Food 36. Kaiko RF, Wallenstein SL, Rogers AG et al. and Drug Administration, 2004. Analgesic and mood effects of heroin and mor- 21. Solomon SD, McMurray JJV, Pfeffer MA et al. the phine in cancer patients with postoperative pain. Adenoma Prevention with Celecoxib (APC) Study N Engl J Med 1981; 304: 1501–1505. Investigators. Cardiovascular risk associated with 37. Zhao SZ, Chung F, Hanna DB et al. Dose-response celecoxib in a clinical trial for colorectal relationship between opioid use and adverse 204 Chapter 15 • Pain control in renal impairment

effects after ambulatory surgery. J Pain Symptom 50. Troy SM, Schultz RW, Parker BD et al. The effect Manage 2004; 28: 35–46. of renal disease on the disposition of venlafaxine. 38. Chaney MA. of intrathecal and epi- Clin Pharmacol Ther 1994; 56: 14–21. dural opioids. Can J Anaesthesiol 1995; 42: 51. Backonja MM. Use of anticonvulsants for treat- 891–903. ment of neuropathic pain. Neurology 2002; 59: 39. Hocking G, Cousins MJ. Ketamine in chronic S14–S17. pain management: an evidence-based review. 52. Jupert P, Almirall J, Casonovas A, Garcia M. Anesth Analg 2003; 97: 1730–1739. Carbamazepine-induced acute renal failure. 40. Weinbroum AA. A single small dose of post- Neurology 1993; 43: 446–447. operative ketamine provides rapid and sustained 53. Bone M, Critchley P, Buggy DJ. Gabapentin in improvement in morphine analgesia in the pres- postamputation phantom limb pain: a random- ence of morphine-resistance pain. Anesth Analg ized, double-blind, placebo-controlled, cross-over 2003; 96: 789–795. study. Reg Anesth Pain Med 2002; 27: 481–486. 41. Collins SL, Moore RA, McQuay HJ et al. 54. Neurontin summary of product characteristics. Antidepressants and anticonvulsants for diabetic Pfizer Ltd, 2005. neuropathy and postherpetic neuralgia: a quant- 55. Lamictal summary of product characteristics. itative systematic review. J Pain Symptom Manage GlaxoSmithKline, 2005. 2000; 20: 449–58. 56. Lyrica summary of product characteristics. Pfizer 42. McQuay HJ. Neuropathic pain: evidence matters. Ltd, 2006. Eur J Pain 2002; 6(A): 11–18. 57. Moore A, Edwards J, Barden J et al. Bandolier’s 43. Sindrup SH, Bjerre U, Dejgaard A et al. The select- Little Book of Pain. Oxford: Oxford University ive serotonin reuptake inhibitor citalopram Press, 2003: 14–18. relieves the symptoms of diabetic neuropathy. 58. Epilim summary of product characteristics, Clin Pharmacol Ther 1992; 52: 547–552. Sanofi-Aventis, 2005. 44. Cymbalta summary of product characteristics. Eli 59. Kalso E, Tramèr MR, McQuay HJ et al. Systemic Lilly and Company Ltd, 2006. local-anaesthetic-type drugs in chronic pain: a 45. Theesen KA, Marsh WR. Relief of diabetic neuro- systematic review. Eur J Pain 1998; 2: 3–14. pathy with fluoxetine. DICP 1989; 23: 572–574. 60. Murphy EJ. Acute pain management pharmaco- 46. Brannon GE, Stone KD: The use of mirtazapine in logy for the patient with concurrent renal or a patient with chronic pain. J Pain Symptom hepatic disease. Anaesth Intens Care 2005; 33: Manage 1999; 18: 382–385. 311–322. 47. Sindrup SH, Grodum E, Gram LF, Beck-Nielsen H. Concentration-response relationship in paroxe- tine treatment of diabetic neuropathy symptoms: a patient-blinded dose-escalation study. Ther Drug Further reading Monit 1991; 13: 408–414. 48. Kiayias JA, Vlachou ED, Lakka-Papadodima E. Australian and New Zealand College of Anaesthetists Venlafaxine HCl in the treatment of painful and Faculty of Pain Medicine. Acute Pain Manage- peripheral diabetic neuropathy. Diabetes Care ment: Scientific Evidence. Australian and New 2000; 23: 699. Zealand College of Anaesthetists, 2005. 49. Davis JL, Smith RL. Painful peripheral diabetic Murphy EJ. Acute pain management pharmacology neuropathy treated with venlafaxine HCl for the patient with concurrent renal or hepatic extended release capsule. Diabetes Care 1999; 22: disease. Anaesth Intensive Care 2005; 33: 311–322. 1909. 16

Diabetes management in kidney disease

Mrudula Patel

Diabetes mellitus is a group of metabolic dis- and develops in 40% of type 1 diabetes mellitus orders characterised by chronic hyperglycaemia and 5–40% of type 2 diabetes mellitus.3 This and is associated with disturbances of carbo- chapter discusses the issues around diabetic hydrate, fat and protein metabolism resulting in renal disease by reviewing the following areas: defects in insulin secretion, action or both. In • The definitions of the various stages of dia- type 1 diabetes mellitus, or insulin-dependent betic renal disease diabetes mellitus (IDDM), there is usually gross • The actual structural changes that occur in destruction of pancreatic beta cells (which pro- the diabetic kidney duce insulin) while in type 2 diabetes mellitus, • The diagnosis of diabetic renal disease or non-insulin-dependent diabetes mellitus • The prevention of diabetic renal disease (NIDDM), insufficient insulin is produced to • The screening and referral process for dia- meet the metabolic needs. Type 2 diabetes betic renal disease mellitus usually occurs later in life than type 1. • The management of diabetes in renal disease. Poorly controlled diabetes mellitus (hyper- glycaemia, hypertension and dyslipidaemia are the key risks) can lead to serious and life- threatening long-term complications referred to Definitions of diabetic renal disease as microvascular and macrovsacular complica- tions. Microvascular complications arise from a cumulative damage to small blood vessels and Diabetic renal disease is a clinical condition include retinopathy, nephropathy and neuro- of altered renal function accompanied by pathy. Macrovascular complications are the structural changes in the kidney. It progresses cardiovascular consequences of the metabolic through a continuum of hyperfiltration and abnormalities associated with diabetes mellitus renal hypertrophy, increased protein excretion which include coronary heart disease, cerebro- to declining renal function leading to end stage vascular and peripheral vascular disease . renal failure (ESRF). In the UK, the National Service Frameworks for both diabetes (2001) and renal services (2005) have highlighted the importance of Microalbuminuria ensuring tight blood glucose and blood pressure control to prevent patients with diabetes mel- Microalbuminuria is defined as a persistent urin- litus developing microvascular and macro- ary albumin excretion between 30 and 300 mg/ vascular complications and then subsequently day. A good estimate is provided by the urinary the detection and management of these long- albumin:creatinine ratio (ACR), with values term complications.1,2 ≥2.5 mg/mmol in men and ≥3.5 mg/mmol in Diabetic renal disease, often referred to as women indicating microalbuminuria. An equiv- diabetic nephropathy, is the leading cause of alent threshold on a timed urine collection is chronic kidney disease (CKD) in Western Europe approximately 20 µg/min.

205 206 Chapter 16 • Diabetes management in kidney disease

Clinical/overt diabetic nephropathy this renal damage there are hormonal changes occurring within the kidney that lead to raised Clinical/overt diabetic nephropathy is defined blood pressure (BP) and this appears to amplify by a persistent urinary albumin excretion the damage process and the amount of protein- ≥300 mg/24 hours and corresponds to clinical uria. At the microalbuminuria stage, sometimes proteinuria as reliably detected by albumin dip- referred to as incipient nephropathy, the renal stick. An equivalent ACR on a random urine function is normal but it should trigger a warn- sample is about 25 mg/mmol.4 ing that the pathological process has started.4

The actual structural changes that The diagnosis of diabetic renal occur in the diabetic kidney disease

The structural changes that occur in the kidney A renal biopsy together with the classical clin- are a continuum, as summarised in Table 16.1. ical features should confirm the diagnosis of There is an initial basement membrane thick- diabetic renal disease. The classical clinical ening of the glomerulus leading to altered pres- features of established diabetic renal disease are: sures and function within the glomerular • At least 5–10 years of diabetes mellitus, this capillaries thus causing a leakage of proteins, includes the undiagnosed period particularly albumin. In the early stages many • Continuous significant proteinuria of the abnormalities are reversible, but eventu- • Mild–moderate increase in BP ally this increasing glomerular damage will lead • Rarely any other findings in the urine (haem- to scarring, permanent loss of function and aturia, cells and casts are all uncommon) dramatically increasing proteinuria. Alongside • Normal or large size kidneys on ultrasound • Usually significant retinopathy, but not always in type 2 diabetes. Table 16.1 The progressive changes with diabetic renal disease Other causes of renal disease can be present in diabetic patients and sometimes co-exist: Progressive changes Status • Renal disease long preceding diabetes Normal GFR normal (e.g 100 mL/min) • Essential hypertension BP normal • Renal stone disease Well • Gout Albuminuria <20 mg/day • Congenital abnormalities • Glomerulonephritis Hyperfiltration Increased GFR (e.g • Pyelonephritis. 120 mL/min) Microalbuminuria Albumin excretion These should be excluded by basic investiga- 30–300 mg/day tions and then treated. Patients with a single Slight increase in BP kidney appear to be more vulnerable to diabetic Persistent proteinuria Progressive increase in renal disease. proteinuria, albumin >300 mg/day Pronounced increase in BP The prevention of diabetic renal Decrease in GFR disease Raised serum creatinine but within normal range Three landmark studies, one in type 1 diabetes, Adapted from ref. 4. DCCT (Diabetes Control and Complication The management of diabetes in kidney disease 207

Trial), and two in type 2 diabetes, UKPDS (UK have the highest risk of mortality and morbid- Prospective Diabetes Study) and the Kumamoto ity of any subgroup in diabetes mellitus. study have all proven the substantial benefits In patients with type 1 and type 2 diabetes, of tight blood glucose control by showing a measure the urinary ACR or albumin concen- reduction in the risk of developing micro- tration annually. If microalbuminuria or pro- albuminuria, proteinuria and rising serum creat- teinuria is present then repeat the ACR or inine.5–7 Epidemiological analysis of the UKPDS albumin concentration twice more within one cohort suggested that with every 1% reduction month where possible. Serum creatinine is in glycated haemoglobin (HbA1c), the chance of measured annually. A referral to a nephrologist developing nephropathy and retinopathy was is made for an opinion when the results are reduced by 35%. The UKPDS also showed abnormal, together with a serum creatinine that by reducing blood pressure (154/87 to greater than 150 µmol/L. Controversy still 144/82 mmHg), there was an associated remains around the standardisation of creatin- decreased risk for developing microalbuminuria. ine measurements to determine renal function. The epidemiological analysis suggested a risk Serum creatinine is significantly lower in dia- reduction of about 12% for each 10 mmHg betes than non-diabetes patients because the decrease in mean systolic BP, with no lower muscle mass is lower. The result will show a bet- threshold. ter renal function but this may be incorrect as All patients who have microalbuminuria or the severity of renal disease in diabetes is under- proteinuria should be started on either an estimated. The American Diabetes Association angiotensin-converting enzyme (ACE) inhibitor has now recommended that physicians use the or an angiotensin II receptor antagonists, and Levey modification of the Cockcroft and Gault this has shown to reduce the rate of progression equation, also known as MDRD (see Chapter 2), to both diabetic nephropathy and non-diabetic to calculate an estimated glomerular filtration nephropathy even when the BP is not obvi- rate (eGFR) from serum creatinine, or of course ously raised.8 Serum creatinine and potassium the original Cockcroft and Gault equation, to should be checked before and after initiating stage the patient’s renal disease. See Chapter 2 treatment or after a dose increase. A small rise for equations. A referral should be made to the in serum creatinine is usually expected in the nephrologist when the eGFR has fallen to short term and should not be an indication to <60 mL/min/1.73 m2 (stage 3 CKD). This high- withdraw therapy, although renal artery sten- lights the fact that patients with stage 3 CKD osis should be considered if a marked creatinine often show a ‘normal’ serum creatinine and rise is seen. 42% will have a serum creatinine <120 µmol/L. The major causes of death in type 2 diabetes In other words, just looking at the serum creat- are the cardiovascular-related illnesses and inine alone, without calculating the patient’s because of this high risk of macrovascular GFR, the CKD would remain undiagnosed. See disease and premature mortality with worsening Chapter 2 for stages of CKD. renal disease, all patients with microalbumin- Once a referral is made to a nephrologist, uria and clinical diabetic nephropathy should long-term management plans can be made to start treatment with a low-dose aspirin and a optimise treatment both in the primary and lipid-lowering agent for vascular protection.9 secondary care setting, thus preventing any fur- ther life-threatening complications.

The screening and referral process for diabetic renal disease The management of diabetes in kidney disease Patients with diabetic renal disease are fre- quently referred too late to initiate treatment to The management of diabetes in kidney disease reduce renal disease progression. These patients should follow the same pathway as the man- 208 Chapter 16 • Diabetes management in kidney disease agement of diabetes mellitus, however consider- alone, in all other treatment options for type 2 ations, and possibly adjustments, should be diabetes. made to account for any renal impairment. For each individual, the potential benefits of The aim of treatment in diabetes mellitus intensive glycaemic control should be balanced is to maintain effective glycaemic and BP con- against the actual or expected risks. trol. The treatment plan for type 2 diabetes should be: Lifestyle interventions for type 2 diabetes • Diet and exercise • Oral monotherapy In type 2 diabetes, lifestyle interventions are key • Oral combination therapy in the prevention and treatment of the condi- • Insulin ± oral agents. tion. The aims are to encourage weight loss, In type 1 diabetes, insulin therapy should be improve glycaemic control and improve the optimised to keep HbA1c as low as possible, cardiac risk factors and potential outcomes by ideally between 6.5% and 7% (the normal level encouraging patients to increase the proportion for healthy individuals is <6%). of vegetables, fruit, nuts and oily fish in their The major limitation of using intensive treat- diet, although patients with ESRF need to be ments in both type 1 and type 2 diabetes is the cautious and avoid all foods that may increase concern of hypoglycaemia, and this risk potassium and phosphate, for example bananas increases as HbA1c is driven down. The fre- and dairy products. quency of hypoglycaemia at any given HbA1c is Dietary interventions should always be the much greater in type 1 compared with type 2 first-line therapy in type 2 diabetes so patients diabetes, although hypoglycaemia does not do not receive the message that the major treat- occur when metformin or the thiazolidine- ment is tablets. In the UKPDS study, dietary diones (or sometimes referred to as the glita- therapy was the sole intervention for the first zones) are used alone, and the risk is lower with three months and achieved a mean 2% decrease the modern insulin regimens, human insulin in HbA1c, more than that achieved by any analogues. Despite this concern of hypogly- single oral hypoglycaemic agent. When the ini- caemia with tight blood glucose control, the tar- tial blood glucose level is high, and the patient get for HbA1c should still be as low as possible, is not highly symptomatic, it is usually better to as long as hypoglycaemia is not a major prob- start with diet and exercise alone, except if there lem. Table 16.2 is a guide for the HbA1c targets. are important personal indications (exams, for- Weight gain is also a common adverse out- eign travel for example), as the sole removal of come of intensive insulin treatment in type 1 large quantities of sugary drinks and simple diabetes, and apart from diet or metformin sugars often leads to a rapid fall in blood glu- cose and control of the symptoms. In those patients with a persistent high blood glucose at diagnosis, then an oral hypoglycaemic agent Table 16.2 Suggested HbA1c targets together with diet should be started. Other Patient group Approximate Action if lifestyle interventions would include exercise target (need to counsel patients on hypoglycaemia with exercise and the undertaking of blood glu- Diet alone, metformin <6.5% >6.8–7% cose monitoring during exercise), stopping or glitazones smoking and reducing alcohol consumption. Sulfonylurea or <7% >7.5% meglitinides Insulin alone or in <7.5% ≥8% Oral hypoglycaemic agents for type 2 diabetes combination As discussed earlier, if the blood glucose level is Adapted from ref. 4. still inadequately controlled, despite the life- The management of diabetes in kidney disease 209

Sites of action for oral therapies for type 2 diabetes

Pancreas ȆSulfonylureas Glucose Gut • Glipizide Impaired • Glyburide insulin secretion • Glimepiride ȆRepaglinide ȇα-Glucosidase inhibitors ȆNateglinide Acarbose Miglitol Hyperglycaemia Pramlintide Exenatide

ȆHepatic Liver glucose ȇGlucose output uptake Muscle Adipose ȇBiguanide ȆTZDs ȇTZDs ȆBiguanide Pioglitazone Metformin

Figure 16.1 Sites of action for oral therapies for type 2 diabetes mellitus. From ref. 10. style interventions, then an oral hypoglycaemic together with flatulence, abdominal pain and a agent (OHA) should be commenced.10 Figure metallic taste. These effects are usually dose 16.1 highlights the sites of action for the vari- related and so always start the patient with a ous OHAs. low dose, 500 mg twice a day, then increase over weeks. Always counsel the patients to take met- formin with meals and never on an empty Metformin (biguanide) stomach and warn patients about the gastro- intestinal side-effects. If the side-effects persist, About 85–95% of newly diagnosed patients with halve the dose and then gradually increase but type 2 diabetes in Western countries are over- more slowly. An emphasis should be made to weight or obese and so the first-line oral agent patients that it will take some days to weeks to is metformin (biguanide) unless it is contra- become effective. The target for most patients is indicated and this will be discussed later. 1000 mg twice a day. Metformin targets the main problem of insulin The contraindications for metformin are con- resistance, and in the UKPDS, the obese group troversial,11 and the main anxiety is lactic acid- treated with metformin showed a much greater osis. Lactic acidosis associated with metformin drop in vascular events than the sulfonylurea/ is a rare condition but it should be withdrawn: insulin groups. Metformin is not associated with weight gain whereas all the other OHAs are • If serum creatinine is higher than 150 µmol/L (the exception are the thiazolidinediones). The (this is a chosen cut-off point for renal fail- mechanism of action of metformin is not fully ure so additional considerations for patients’ understood, but it does decrease hepatic gluco- muscle mass and protein turnover should be neogenesis and enhances peripheral glucose made) utilisation and thus reduces appetite. It is only • During periods of suspected tissue hypoxia effective if there are some residual functioning (sepsis, myocardial infarction. etc.) pancreatic beta cells. The main drawback of • Three days after contrast medium containing metformin is the high incidence of gastro- iodine has been given, and restart metformin intestinal side-effects, nausea and diarrhoea, only when renal function has been checked 210 Chapter 16 • Diabetes management in kidney disease

• Two days before general anaesthesia and Otherwise they are well tolerated with occa- restart only when renal function is stable. sional effects of rashes, nausea, vomiting, dia- rrhoea and constipation. They may rarely cause In diabetic kidney disease, metformin is still hepatic problems including cholestatic jaundice the first-line agent unless the eGFR falls below and eventual liver failure so are contraindicated 60 mL/min/1.73 m2 as discussed above. In an in severe hepatic disease. Blood dyscrasias and acute episode of renal failure, metformin should severe skin rashes have also been seen rarely. be stopped but can be safely restarted if the They are contraindicated whilst breastfeeding eGFR returns to above 60 mL/min/1.73 m2 and and in patients with porphyria. the renal function routinely measured thereafter, Patients should always be taught how to if not an alternate OHA, usually a sulfonylurea, recognise hypoglycaemia and told weight gain should be commenced. Other contraindications is expected. There is no benefit at all in exceed- include pregnancy or breastfeeding. ing the maximum dose or using more than one sulfonylurea, so another group of OHAs should Sulfonylureas be added, usually metformin, unless it is contra- indicated, in which case a glitazone is a good Sulfonylureas increase pancreatic insulin secre- choice. tion, therefore are only effective when there is some residual pancreatic beta cell activity. The Thiazolidinediones (glitazones) most commonly prescribed sulfonylureas are gliclazide, glipizide, glimepiride and gliben- The thiazolidinediones, often referred to as the clamide; less commonly seen are chlor- glitazones, are selective agonists at the peroxi- propamide and tolbutamide. Hypoglycaemia is somal proliferator-activated receptor gamma the main issue with sulfonylureas, especially in (PPARγ) nuclear receptor and reduce glycaemia renal impairment, and sulfonylurea-induced by reducing insulin resistance at adipose tissue, hypoglycaemia may persist for many hours, skeletal muscle and liver. The glitazones have a often requiring hospitalisation. The long-acting synergistic action with other OHAs, so the sulfonylureas chlorpropamide and gliben- National Institute for Health and Clinical clamide are associated with a higher risk of Excellence (NICE) in the UK have sanctioned hypoglycaemia. their use as second-line therapy with either met- Chlorpropamide is contraindicated in severe formin or a sulfonylurea, where the glitazone renal impairment, eGFR <20 mL/min/1.73 m2, should replace whichever drug in the combina- but can be used with caution if the eGFR is tion is poorly tolerated or contraindicated. between 20 and 50 mL/min, and as well as the At present there are only two glitazones increased risk of hypoglycaemia, there is a available for the management of type 2 dia- severe risk of metabolic acidosis. Glibenclamide betes, pioglitazone and rosiglitazone, both of in renal impairment, again should be used with which were licensed in the UK in 2000. The two caution and initially at low doses, 1.25–2.5 mg landmark studies for patients with type 2 dia- once a day with monitoring. The short-acting betes, UKPDS (1998) and Kumamato (1995), sulfonylurea gliclazide is the usual drug of occurred well before the glitazones were manu- choice in renal impairment, and doses start factured, thus publication data on outcome between 20 mg and 40 mg once a day (refer to results for their use have been fairly limited. In the Renal Drug Handbook for further drug dosing general the glitazones produce similar glycaemic information12). Again, as with renal impair- improvements (1–1.5% HbA1c) to other OHAs ment, sulfonylureas should be used with cau- either as monotherapy or as combined therapy, tion in elderly or frail patients or those living although as mentioned earlier, NICE guidelines alone, as severe protracted hypoglycaemia can restrict their use for dual therapy only. From the occur. limited evidence available, they have been Other side-effects include weight gain, which shown to maintain effect in the long term and is common especially in the first 3–6 months. preserve beta cell function, have beneficial The management of diabetes in kidney disease 211 effects on lipids and many other cardiovascular The meglitinides risk factors. The meglitinides stimulate insulin secretion and Originally there were concerns about hepatic are the newest group of antidiabetics licensed in toxicity, oedema and heart failure. The hepatic the UK (2001). There are two drugs available, toxicity anxiety is probably unfounded and repaglinide and nateglinide. Both drugs have dates back to the first glitazone, troglitazone, different mechanisms of action but pre- that was licensed in the United States. Now dominately enhance first-phase insulin release, there is increasing evidence that the glitazones and therefore reduce immediate post-prandial may benefit fatty liver, although the manufac- hyperglycaemia. New patients should always be turers still recommend liver function tests (LFTs) initiated on a small dose and then titrated should be carried out before therapy and every according to blood glucose. two months for the first year and periodically Repaglinide has been licensed as mono- thereafter. The remaining side-effects are weight therapy or in combination with metformin, gain and oedema. There is concern about pre- while nateglinide has a licence for combination cipitation of heart failure, especially in patients with metformin only. Both drugs have a rapid on insulin, so the glitazones at present should onset of action, so patients should be advised to not be used in patients with heart failure or at take the tablet up to 30 minutes prior to meals. a very high risk for it. They are also contra- The duration of activity is short, so omit the indicated for co-administration with insulin, dose if a meal is missed. during pregnancy and while breastfeeding. Had the meglitinides been invented before The glitazones are extensively metabolised in the sulfonylureas and were less expensive, they the liver, some of the metabolites are active, and would probably be preferred as they cause less are excreted mainly via the urine and 25% via hypoglycaemia and minimal weight gain, but faecal elimination. When using the glitazones they are more expensive, and this has limited in diabetic kidney disease, therefore, no dose their use. adjustment is required if patients have mild to The meglitinides in diabetic kidney disease moderate renal impairment. The data for their require no dose adjustments in patients with use in ESRF are limited and the manufacturers renal impairment and as nateglinide is licensed recommend that the glitazones should be used only in combination with metformin, it would with caution. In practice, the glitazones can be not be considered for patients with ESRF. Side- used at normal doses in ESRF, but blood glucose effects include gastrointestinal upsets and rashes should be monitored especially during any and rarely hypoglycaemia. They are also contra- acute episodes (refer to the Renal Drug Handbook indicated in severe hepatic impairment, during for further drug dosing information12). pregnancy and while breastfeeding. The glitazones take several months to show their full effect (up to six months), so doses should not be rapidly adjusted and patients Acarbose should be encouraged to continue treatment. Acarbose is an inhibitor of alpha-glucosidase, Some consultant diabetologists are trialling and therefore limits and delays the absorption triple therapy (metformin + sulfonylurea + glita- of starches. It has a mild hypoglycaemic effect zone) for a 2–3 month period to account for this with a mean HbA1c reduction of around 0.5% delayed effect from the glitazones and then but its gastrointestinal side-effects, mainly flat- stopping whichever initial drug (metformin or ulence, makes it an unpopular choice. However, sulfonylurea) was ineffective. Around 25% of it may have a small role in patients unable to patients will show no blood glucose response to tolerate metformin and in combination therapy. the glitazones; such patients tend to be more obese and have a long-standing insulin resist- Combination therapy for type 2 diabetes ance with depleted pancreatic insulin reserves. In those non-responders there should not be a If monotherapy with a single OHA fails to con- delay to move over to insulin. trol blood glucose, then a second OHA should 212 Chapter 16 • Diabetes management in kidney disease be added. The most common combination for Insulin therapy in type 1 diabetes type 2 diabetes is metformin with a sulfonyl- Most patients with type 1 diabetes continue to urea, but if this combination is contraindicated, produce their own insulin for a year or so then the alternate options could be: after diagnosis, which becomes apparent soon after starting insulin as the removal of glucose • Metformin with a glitazone toxicity allows the pancreatic beta cells to • Sulfonylurea with a glitazone recover, resulting in a reduction in the total • Metformin with a sulfonylurea and a amount of insulin required, sometimes referred glitazone (triple therapy, not ideal for the to as the ‘honeymoon period’, lasting 6–12 long term and insulin would be a better months. alternative) • Metformin with a meglitinide Insulin therapy in type 1 and type 2 diabetes • Metformin with acarbose. The aim of insulin treatment in both type 1 and However, most people with type 2 diabetes type 2 diabetes is to maintain effective gly- will eventually progress to needing insulin due caemic and BP control. to pancreatic beta cell exhaustion, and it is Ideally, insulin regimens should mimic as important to identify these patients promptly, closely as possible the insulin profile of healthy thus preventing microvascular and macro- individuals, a prandial bolus insulin with a basal vascular complications. insulin, often referred to as a basal–bolus regi- men (i.e. rapid/short-acting insulin to control postprandial spikes with a long-acting insulin to Insulin give a low-level background cover). The recent advances in insulin technology has led to the Insulin therapy in type 2 diabetes introduction of new human insulin analogues, Insulin therapy, as mentioned earlier, should be thus revolutionising insulin treatment for dia- initiated as promptly as possible if OHAs cannot betes and allowing specific tailoring for each achieve HbA1c <8%. If there is still some resid- individual.13 ual insulin secretion, then patients with type 2 The insulin groups now available are: diabetes will usually administer insulin at night • Rapid-acting analogues (immediate or prolonged action) and continue • Short-acting with a daytime OHA (usually metformin or a • Intermediate-acting sulfonylurea). This use of a nocturnal insulin • Long-acting will suppress hepatic glucose production • Prolonged-action analogue. overnight, therefore controlling the fasting blood glucose levels, whilst continuing a day- Insulin therapy in diabetic kidney disease time OHA will minimise the weight gain. The Insulin in a healthy individual circulates free as new prolonged action basal insulin analogues a monomer with a half-life of 4–5 minutes. It is glargine and detemir are increasingly being used metabolised by the liver and kidney. In the kid- to cover the overnight insulin as clinical trials ney, insulin is filtered by the glomeruli and have proven a reduction in nocturnal hypo- reabsorbed by the tubules which also degrade it. glycaemia and weight gain compared with the In renal and hepatic disease, there is a decrease older intermediate insulin. in the rate of insulin clearance, resulting in For those patients with type 2 diabetes where increased amounts of circulating insulin, thus there is complete pancreatic beta cell exhaus- increasing the risk of hypoglycaemia, so insulin tion insulin alone will be the only option. These doses should be reduced. In diabetic kidney dis- patients will need to be carefully monitored and ease this reduction in insulin requirement will the dose adjusted in order to balance and pre- vary depending on the degree of renal impair- vent weight gain and hypoglycaemia from the ment. In ESRF, the insulin dosing may need a insulin regimen. The management of diabetes in kidney disease 213 further reduction as there is a complete failure of inhaled insulin, but the current evidence has of the kidney to remove insulin, together with shown that it is as effective as injectable insulin the anorexia and weight loss associated with in blood glucose control in type 1 diabetes but uraemia, which can lead to virtual starvation. the dosing regimen is complicated and wasteful Some patients in ESRF even manage to come off and the equipment is not convenient. treatment completely.14 At present there are no published data for its For those patients in ESRF the type of renal use in renal impairment.16 replacement therapy, haemodialysis or peri- toneal dialysis, will also decide the type and Rapid-acting insulin analogue, glulisine dose of insulin required. Haemodialysis usually (Apidra) takes place three times a week for around 4 The rapid-acting insulin analogue glulisine hours each session so this will interrupt the (Apidra) has been introduced and licensed for daily eating habit as well as the daily activity for use in both type 1 and type 2 diabetes. Evidence these patients. Also, because insulin is a big to date has shown its advantage as an alternat- molecule, it will not be removed by haemo- ive for obese or overweight patients.10 dialysis, thus increasing the incidence of hypo- glycaemia. The options for the patient on Glucagon-like peptide 1 analogue (Exenatide) haemodialysis are to reduce the dose of insulin The glucagon-like peptide 1 (GLP-1) analogue on haemodialysis days or to use a once-a-day Exenatide has been available in the USA since prolonged-acting basal insulin for a low-level 2005 but is awaiting a European regulatory 24-hour background cover. review. Exenatide is an injection and a member Peritoneal dialysis occurs on a daily basis, at of a new class of drugs which mimic the set times and is less aggressive. It will therefore glucose-lowering action of human hormones have a minimal effect on the overall glycaemic called incretins. In the USA, it has been control, although the peritoneal dialysis approved as adjunctive therapy for patients dialysate will contain glucose and its absorption with type 2 diabetes with metformin or a can increase the blood glucose levels in patients sulfonylurea, or both. There are no published on PD. Hence these patients generally require data for its use in renal impairment.17 higher doses of insulin than haemodialysis patients.15 Islet cell transplantation Islet cell transplantation is currently available in some hospitals around the UK as a potential New agents cure and or change to the natural history of the disease diabetes. Inhaled insulin (Exubera) The inhaled insulin Exubera has recently received Pancreas transplantation a licence in the UK for certain patients with type Pancreas transplantation is a solid organ trans- 1 and type 2 diabetes. It is fast acting and when plant and a potential cure/change for diabetes. inhaled will reach the lungs within 10 minutes Again, as with islet cell transplantation, it is an before starting a meal. NICE are in the process option available in some hospitals around of formalising a technology guidance on the use the UK. 214 Chapter 16 • Diabetes management in kidney disease

C CASE STUDY

DP is a 76-year-old woman, weighing 99.1 kg, who presents with acute renal failure, shortness of breath, tachycardia and diarrhoea. She has a past medical history of hypertension, type 2 diabetes mellitus, chronic renal failure (diabetic nephropathy) and has recently been treated for cellulitis with flucloxacillin then cefalexin by her GP.

Drug history: • Metformin 850 mg twice daily (stopped on admission) • Gliclazide 160 mg every morning 80 mg every night (withheld) • Rosiglitazone 4 mg every morning (withheld on admission) • Atenolol 100 mg every morning (stopped on admission) • Valsartan 40 mg every morning (stopped on admission) • Atorvastatin 40 mg every night (stopped on admission) • Aspirin 75 mg every morning (stopped on admission).

Family history: Nil relevant.

Social history: Lives alone and independent.

Biochemistry:

Day 1234101112

Creatinine (µmol/L) 716 641 454 319 248 204 192 Urea (mmol/L) 49.2 34.3 35.2 30.2 25 23 23 Sodium (mmol/L) 136 139 137 138 137 137 134 Potassium (mmol/L) 7.1 5.7 4.8 4.5 3.0 3.2 3.2 Bicarbonate (mmol/L) 17 18 22 22 24 25 24 Calcium (mmol/L) 1.98 2.14 2.34 2.45 2.35 2.24 2.24 Phosphate (mmol/L) 2.39 2.54 1.64 1.38 1.07 0.89 0.90 Alb (g/L) 38 38 43 43 39 38 38 BP (mmHg) 70/40 85/31 110/50 115/50 130/75 120/80 120/75 Blood glucose (7 am) 4.5 6.2 7.7 7.6 10.1 12 5.3 (mmol/L) Blood glucose (noon) 8.5 12.9 11 14.7 10.1 23.5 12 (mmol/L) Blood glucose (10 pm) 8.7 8.2 7.4 16 12.2 13.2 14 (mmol/L) HbA1c (%) 9.0 Hb g/dL 7.6 10.9 11.0 12.3 12.3 12 11 WCC (109/L)12999999 Weight (kg) 99.1 94.4 92.5 91.1 90 90 90 → The management of diabetes in kidney disease 215

C CASE STUDY (continued)

The main issues are the acute renal failure (anuric, hyperkalaemic, acidotic), shortness of breath and fluid overloaded, hypotension, tachycardia, diarrhoea and poor blood glucose control. On day 1, patient DP is haemofiltered. Metformin, valsartan and atenolol are stopped and all other medicines are withheld. Ranitidine is commenced temporarily for uraemia. A chest X-ray shows pulmonary oedema but no signs of chest infection. Cultures are taken to rule out sepsis. Patient has signs of peripheral oedema. She is fluid restricted and furosemide 120 mg every morn- ing is started. Tropium level is taken to rule out any cardiac event; the result is <0.2 µgram/L. On day 2, DP is haemofiltered again although she has started to pass some urine and the peripheral oedema is improving. The diarrhoea has stopped. On day 3, the patient has passed >2 L of urine and her biochemistry is improving. Blood cultures return as no growth; patient is apyrexial, C-reactive protein is returning towards normal. Blood glucose is reviewed, but as the patient is not eating much, she remains solely on gliclazide 80 mg every morning to control blood glucose. Her weight is down to 92.5 kg. Renal USS shows no new abnormalities. On day 4, the patient continues to improve, passing good urine and eating well. Atorvastatin and aspirin are restarted. On day 10, the patient is reviewed by the diabetologist for blood glucose control, and insulin, Novomix 30 twice daily, is recommended and oral antidiabetics are stopped. On day 11, valsartan 40 mg every morning is restarted as results show no renovascular disease. On day 12, the patient is responding well to insulin and discharge plans are made.

Q1. What might her causes of diabetes be?

Q2. What might her causes of chronic renal failure be?

Q3. Could it have been prevented?

Q4. What might her causes of acute renal failure be?

Q5. Explain her drug history and any possible reasons for stopping some of her medication on admission.

Q6. Why would this patient take atorvastatin and aspirin?

Q7. Why change this patient’s diabetic regimen to insulin?

Q8. Would the regimen differ if the patient had ESRF?

Q9. Comment on BP control in diabetes.

Q10. What discharge plans should be made for this patient? 216 Chapter 16 • Diabetes management in kidney disease

References mellitus: a randomised prospective 6-year study. (Kumamoto Study). Diabetes Res Clin Pract 1995; 28: 103–117. 1. Department of Health. National Service Framework 8. Mogensen CE, Cooper ME. Diabetic renal disease: for Renal Services, Part Two: Chronic Kidney Disease, from recent studies to improved clinical practice. Acute renal Failure and End of Life Care. London: Diabet Med 2004; 21: 4–17. Department of Health, 2005. 9. National Prescribing Centre NHS. Drug manage- 2. Department of Health. National Service Framework ment of type 2 diabetes: summary. MeRec Bull for Diabetes: Standards. London: Department of 2004; 15. Health. 2002. 10. Medscape. Diabetes and insulin: Indications, 3. Hasslacher C. Diabetic Nephropathy. London: initiation and innovations. http//medcape.com/ Wiley, 2001: 22–27. viewprogram/5500_pnt (accessed 13 June 2006). 4. Drury P, Gatling W. Your Questions Answered. 11. Alexander WD, Jones GC, Macklin JP. Contra- Diabetes. London: Churchill Livingstone, 2005. indications to the use of metformin. BMJ 2003; 5. The Diabetes Control and Complications Trial 326: 4–5. Research Group. The effect of intensive treatment 12. Ashley C, Currie A. The Renal Drug Handbook, 2nd of diabetes on the development and progression edn. Oxford: Radcliffe Medical Press, 2004. of long-term complications in insulin-dependent 13. Gummerson I. An update on insulin analogues. diabetes mellitus. N Engl J Med 1993; 329: 977– Pharm J 2006; 277: 169–172. 986. 14. Bilous RW. End-stage renal failure and manage- 6. Alder AI, Stevens RJ, Manley SE et al. ment of diabetes. Diabet Med 2004; 21: 12–14. Development and progression of nephropathy in 15. Atherton G. Specialism within a specialty: dia- type 2 diabetes: The United Kingdom Prospective betes and dialysis. Br J Renal Med 2003/2004; 8: Diabetes Study (UKPDS 64). Kidney Int 2003; 63: 10–11. 225–232. 16. Barnett EJ, Morton-Eggleston E. Inhaled insulin. 7. Ohkubo Y, Kishikawa H, Araki E et al. Intensive BMJ 2006; 332: 1043–1044. insulin therapy prevents the progression of dia- 17. Dungan K. Treating diabetes by modifying GLP-1 betic microvascular complications in Japanese activity: Current options and new developments. patients with non-insulin-dependent diabetes Diabet Microvasc Complic 2006; 3: 43–46. 17

Myeloma and kidney disease

Elizabeth Lamerton

Myeloma is a cancer affecting plasma cells or result comparison of data is somewhat unreli- white blood cells in the bone marrow. The able. As an approximate guide, 43% of 998 cancerous plasma cells proliferate and fill more patients in one study had a creatinine greater than 10% of the bone marrow, preventing the than 133 µmol/L and 22% of 423 patients had a formation of normal blood cells. Patients with creatinine greater than 177 µmol/L.2,3 Advanced myeloma may present with a range of non- acute renal failure (ARF) requiring urgent dia- specific symptoms including kidney disease, lysis therapy occurs in 3–12% of all cases.4 anaemia and infections. Myeloma and kidney disease are undoubt- Myeloma cells produce an abnormal anti- edly closely linked, with up to 30% of all body protein known as paraprotein. The prolif- myeloma patients having some degree of kidney eration of plasma cells produces an excess of damage at presentation and over 50% develop- the abnormal proteins. In addition, incomplete ing kidney damage during their illness.2 immunoglobulins are produced, for example In myeloma the paraprotein light chains are monoclonal immunoglobulin. filtered in the kidney across the glomerulus, Kidney disease is a common complication of where some will be reabsorbed by the tubular multiple myeloma with approximately 50% cells. Light chains that are incompletely re- of patients having kidney disease at diagnosis. absorbed by tubular cells are found in the urine There are a number of possible mechanisms of as Bence-Jones protein. Urinary Bence-Jones kidney damage and often more than one mech- proteins can be detected and quantified in the anism will be involved. laboratory. Simultaneously, myeloma cast neph- The incidence of multiple myeloma in the ropathy may occur, where immunoglobulin UK and Scandinavian countries is approxim- light chains are deposited in the kidney as ately 50 cases per million.1 The incidence rises casts. In cast nephropathy, the filtered immuno- with progressing age, with a mean of 70 years globulin light chains bind to a common site on (Table 17.1). Tamm-Horsfall protein. Aggregation of the pro- An exact incidence of kidney disease associ- tein complexes produces casts that can cause a ated with multiple myeloma is difficult to physical obstruction in the tubular cells. This quantify. The definitions of kidney disease have can result in kidney disease. varied in the UK and internationally and as a The presence of paraproteins and the deposi- tion of fibrils causes related conditions such as amyloid disease, and crystals of abnormal pro- tein light chains cause light chain deposition Table 17.1 Incidence of myeloma by age disease and Fanconi’s syndrome.1,2 Myeloma kidney is associated with a high Age <40 years <2% initial mortality of approximately 20% within Age 40–60 years 15% one month of referral to a specialist centre. The Age 60–65 years 15% poor survival rates reflect the large tumour General population 50 cases per million burden often already present at diagnosis and is

217 218 Chapter 17 • Myeloma and kidney disease due to both underlying haematological malig- • Quantification of monoclonal protein in nancy and to ARF.5 serum and urine. The presence of any monoclonal protein – m-protein or paraprotein – indicates a plasma cell abnormality and warrants detailed invest- Clinical presentation of multiple igation and referral to a haemato-oncologist.1 myeloma

Patients with multiple myeloma may present in a number of ways. Patients with symptoms of Myeloma kidney disease or organ damage require urgent referral, assessment and treatment. Some may present to Myeloma kidney is a general term that is hospital with an acute medical emergency, such applied to both ARF and to chronic kidney dis- as spinal cord compression, hypercalcaemia or ease associated with multiple myeloma. acute kidney disease, especially ARF. Other patients may present with non-specific clinical signs such as: Acute renal failure associated with myeloma • Bone disease, in particular unexplained chronic back pain without an obvious pre- There are a number of factors responsible for cipitating cause ARF associated with myeloma. In up to 80% of • Normochromic, normocytic anaemia with cases, light chain-induced cast nephropathy is no clear cause responsible and this is worsened by the presence • Recurrent infection of sepsis, hypovolaemia and hypercalcaemia. • Hyperviscosity syndrome For further discussion on contributing factors to • Features of amyloidosis such as nephritic ARF see Chapter 3.2 syndrome and cardiac failure • Raised erythrocyte sedimentation rate (ESR) or plasma viscosity Chronic renal disease associated with myeloma • Chronic kidney disease. The three most common kidney diseases associ- ated with multiple myeloma are: • Cast nephropathy Investigation and diagnosis of myeloma • Primary amyloidosis • Light chain deposition disease. When myeloma is suspected, patients require at They are all associated with overproduction of least the following tests and investigations: monoclonal immunoglobulin light chains. • Full blood count • Erythrocyte sedimentation rate Light chains • Serum and plasma electrolytes • Electrophoresis of serum and concentrated Light chains are not detected on routine urine urine dipstick. A 24-hour urine collection is usually • Quantification of immunoglobulin in urine sent to the biochemistry and immunology lab- and serum oratories for detailed analysis using techniques • Skeletal survey – X-ray, CT or MRI such as immunoelectrophoresis and immuno- • Bone marrow aspirates or trephine biopsy fixation. Light chains have a molecular weight and histological review under the microscope of 22 000 Da and are therefore filtered freely • Immunofixation of serum and urine across the glomerulus before re-absorption in Treatment options 219 the proximal tubular cells. In myeloma the light chain excretion rate is markedly increased Table 17.2 Dose of pamidronate from less than 30 mg/day to 100 mg–20 g in 24 hours. Serum calcium (mmol/L) Dose (mg) The biochemical characteristics of the indi- Up to 3.0 15–30 vidual light chains determine the type of renal 3.0–3.5 30–60 disease observed. Generally patients will exhibit 3.5–4.0 60–90 only one type of light chain-associated kidney >4.0 90 disease at any given time.

Renal tubular dysfunction ments are high and may be given in combina- The glomerular filtration rate (GFR) may be rel- tion with localised therapy such as radiotherapy atively unaffected in some patients where the or surgical intervention. Pain symptoms often direct toxicity from the filtered light chains improve following chemotherapy and associ- affects only the renal tubular cells. The clinical ated disease regression. For detailed discussion consequences of tubular disease in myeloma on pain control in patients with kidney disease may include proximal renal tubular acidosis and see Chapter 15. phosphate losses, both of which are signs of In myeloma patients, bone pain may also be Fanconi’s syndrome. effectively treated with bisphosphonate therapy, and clodronate, pamidronate and zoledronic acid have all been used in both clinical trials Amyloidosis and light chain deposition disease and routine clinical settings.1 See Table 17.2 for dose of pamidronate. The proliferation of plasma cells in myeloma Patients with kidney disease may require leads to an excess of monoclonal light chains modification of both the volume of bisphos- and rarely heavy chains. The monoclonal phonate infusion and a reduction in the rate of chains can fragment and deposit immuno- administration. Intravenous bisphosphonates globulin around the body. This process causes such as zoledronic acid should be used with primary amyloidosis, light chain deposition dis- caution in patients with any stage of kidney ease and heavy chain deposition disease. disease.

Hypercalcaemia Increased bone resorption contributes to the Treatment options presence of hypercalcaemia in 15% of patients with multiple myeloma. Hypercalcaemia alone Treatment of myeloma is not usually curative may worsen ARF and must be treated urgently but aims to control the disease by putting the if it is one of the presenting features (see patient in remission or slowing progression and Chapter 3). to effectively control symptoms. Hypercalcaemia may be treated with ag- Regular monitoring of kidney function is gressive rehydration and intravenous bisphos- essential to ensure speedy identification of phonates. developing kidney damage. Advise high fluid intake to promote good urine output, and allow correction of dehydration. Any abnormal Bone pain electrolytes should be treated appropriately, Pain is a common complaint in patients with including hypercalcaemia. It is important to myeloma due to the common skeletal manifest- avoid any potentially nephrotoxic agents where ations of disease. Systemic analgesia require- possible. 220 Chapter 17 • Myeloma and kidney disease

Therapy of myeloma may be considered in taining a future possibility of stem cell mobil- three stages: induction, maintenance and isation and transplantation. The commonly relapse.1 used regimens include, vincristine, doxorubicin and dexamethasone (VAD) given as a continu- ous infusion for 4 days or a combination of Induction therapy cyclophosphamide, doxorubicin, vincristine, methotrexate and prednisolone. These com- Management of symptoms binations have been shown to offer a higher response rate and cure rate of 10–25%.1 There At presentation, pain control is a common have been no large trials, however, comparing feature and requires immediate attention. Good the different regimens with each other.1,6,7 pain control is usually achieved using a com- Doses may have to be adjusted according to bination of factors: renal function. • Analgesia The infusional regimens have the signific- • Radiotherapy ant disadvantage of requiring central venous • Orthopaedic intervention access, usually through a permanent central • Chemotherapy to control the underlying venous catheter. This is associated with signi- pathological process. ficant infection risks and may be particularly difficult in patients with end stage kidney disease who also require central access for Analgesia haemodialysis. Conventional chemotherapy is given to Systemic analgesic options may be complicated patients where a response is required but by associated kidney disease (see Chapter 15). toxicity should be avoided. This treatment is Alternative analgesic approaches include local generally expected to be less aggressive and radiotherapy for skeletal disease. Myeloma oral treatment is often chosen rather than skeletal metastases have been found to be radio invasive parenteral therapy. One of the most responsive and therefore this may be used for common protocols used in this setting is palliation. melphalan and prednisolone, based upon the conclusions of the Myeloma Trialist’s Radiotherapy Collaborative Group in 1998. Patients must be monitored for signs of toxicity, and particular High doses of radiation may be targeted at dis- attention must be paid to the nadir cell counts eased bone marrow to treat severe localised and duration. Doses require modification for problems. Radiotherapy is often used for both toxicity and in patients with decreased symptom management, for example, as targeted renal function.8 therapy for bone pain caused by myeloma cells Chemotherapy for patients presenting with in the bone marrow. organ damage to the kidneys aims to reduce the quantity of paraprotein and light chains and thereby minimise organ damage. Chemotherapy Chemotherapy will be considered for patients Melphalan with myeloma symptoms and for patients with Melphalan is predominantly excreted by the myeloma-related organ damage, such as kidney and therefore as for conventional ther- myeloma kidney, in the absence of other apy, requires careful monitoring and adjustment myeloma symptoms. for bone marrow suppression. Retrospective In patients for whom high-dose chemo- analysis of the Nordic Myeloma Study Group therapy is planned, initial chemotherapy is (NMSG) trials9 prompted the authors to propose given to reduce the cancer load, while main- a 25% initial dose reduction of melphalan and Treatment options 221 individualisation of doses thereafter according Lenalidomide to bone marrow suppression.1 Lenalidomide has both immunomodulatory and anti-angiogenic properties, with multiple Cyclophosphamide mechanisms for this action. It is rapidly Cylophosphamide is a pre-drug whose meta- absorbed orally and approximately 65% of the bolites are cleared by the kidney. Manufacturers parent drug is excreted unchanged in the urine. recommend dose reductions of 25% in stage 4 Patients with kidney disease may require a dose chronic kidney disease (CKD) and 50% in stage modification to avoid accumulation and should 5 CKD (see Chapter 2 for classification of CKD). be carefully monitored for adverse effects and It is recommended that doses should be toxicity. monitored and adjusted according to effect and toxicity. Bortezomib Bortezomib is a new agent being used in the Dexamethasone therapy of myeloma. It is a proteasome Dexamethasone is often given as single-agent inhibitor. There are on-going clinical trials to initial therapy for patients with myeloma while establish the optimum combination and most future chemotherapy regimens are decided. No efficacious stage at which this therapy should 1 dose reduction is necessary and pulsed high- be given. dose oral doses are given for 4 days.7

Maintenance phase Combination regimens – infusional chemotherapy Chemotherapy may be required for a prolonged Infusional chemotherapy regimens discussed period of time – often at least 12 months until above, such as VAD chemotherapy, may also be the patient’s disease is in the plateau phase, administered to patients with kidney disease with stable M-proteins in serum and urine and without dose modification.6 no clinical or laboratory evidence of progres- sion. The plateau phase describes a period of dis- Thalidomide ease stability of at least 4–6 months following The exact mechanism of action for thalidomide chemotherapy. Maintenance chemotherapy is is uncertain. There are a number of actions: usually not recommended in the plateau phase although some patients will continue to receive • Direct suppression of the growth and survival drugs such as thalidomide long term. of myeloma cells During the maintenance phase of therapy • Direct kill of myeloma cells patients will be monitored for disease relapse • Alteration of the molecules that allow and progression. myeloma cells to attach themselves and grow within the bone marrow • Alteration of the production and activity of chemicals involved in the growth and sur- Relapse of disease vival of myeloma cells When patients relapse and symptoms return, • Inhibition of angiogenesis – growth of new rapid reduction of paraprotein excretion is the blood vessels first aim of therapy. Initial therapy is given • Stimulation of lymphocytes to attack immediately using high dose steroids such as myeloma cells. pulsed dexamethasone 40mg daily for 4 days for Thalidomide undergoes non-enzymatic hyd- a rapid response. rolysis to multiple degradation products and is In the presence of ARF, patients will usually not dependent on renal clearance. It is therefore receive pulsed steroid therapy in addition to not necessary to adjust doses for kidney disease.1 plasmapheresis or plasma exchange to filter 222 Chapter 17 • Myeloma and kidney disease excessive amounts of paraprotein from the Treatment of anaemia blood. Haemodialysis or haemofiltration may be required as a life saving intervention in ARF. Sixty per cent of patients with myeloma will have normochromic, normocytic anaemia at presentation. The incidence of anaemia Treatment of hypercalcaemia increases in patients with recurrent or aggressive progressive disease who have received previous During active myeloma disease, hypercalcaemia chemotherapy.1 may be found in up to 30% of patients. Previously, anaemia was managed with regu- Hypercalcaemia is directly nephrotoxic and lar and repeated red blood cell transfusions. This exacerbates light chain-mediated nephrotoxicity. method of management was associated with sig- Management strategies include adequate nificant transfusion risks, hospital admission hydration and intravenous bisphosphonate time and the additional risk of accentuating therapy. hyperviscosity in patients with high paraprotein Fluid replacement may be complicated in levels. It is therefore preferable to avoid admis- patients with fluid restriction due to kidney sion to hospital for blood transfusions and out- disease. patient alternative therapy has been sought. Parenteral bisphosphonates must be used The growth in the use of erythropoiesis- with caution as they have been associated with stimulating agents (ESA) in the therapy of nephrotoxicity. Consequently bisphosphonates renal anaemia over the past 10 years has led to require reduction in both dose and rate of further investigation and evaluation in the administration in the presence of kidney dis- oncology and haematology settings. There is ease. Individual assessment of the risks and strong evidence for the use of recombinant benefits must be made on each occasion. human erythropoietin in the treatment of chemotherapy-related anaemia in myeloma. The use of ESAs in patients with myeloma is Treatment of kidney disease in multiple recommended by the American Society of Clin- myeloma ical Oncology (ASCO) and the American Society of Hematology (ASH) and the British Society of The MERIT trial is a multicentre study to evalu- Haematologists.1 ate the effectiveness of plasma exchange in patients with newly diagnosed myeloma and Dose of erythropoietin-stimulating agents ARF.10 Plasma exchange has been used to reduce In contrast to patients with anaemia of chronic serum light chain levels and minimise long- kidney disease, the starting doses of ESA for term kidney damage. Plasma exchange is a sim- patients with myeloma is notably higher: epo- ilar process to dialysis, but is significantly more etin beta 30 000 units per week or darbepoetin at removing plasma proteins. 150 µg each week. This dose should be doubled Long-term kidney damage may be reduced by if no improvement in haemoglobin is observed prompt intervention and correction of early after 4–6 weeks. signs of kidney disease. Urine output over 3 L in 24 hours should be maintained where possible, guided by the patient’s fluid status. Central Treatment of infections venous pressure monitoring may be required and is a useful guide to fluid replacement. The combined effects of haematological disease, Hypercalcaemia should be corrected as kidney disease and chemotherapy result in described above with fluids and bisphosphonate immunosuppression and leave patients at a very therapy. high risk of infections. Infections carry an asso- Infection must be aggressively managed as ciated high morbidity and mortality in this sepsis is the commonest cause of mortality in all patient group and must be treated aggressively stages of myeloma kidney disease.1,4 with appropriate antibiotics.1 Summary 223

Summary chemotherapy and in the prevention and treat- ment of associated kidney disease. Perhaps the most important role is ensuring kidney disease The management of multiple myeloma poses a is not exacerbated by myeloma therapies and in challenge for the healthcare professions and for ensuring patients are not unnecessarily exposed patients. The role of the pharmacist is essential to increased adverse effects from medicines. both in the treatment of myeloma with

C CASE STUDY

Mr CB is a 64-year-old man transferred to the renal ward for further investigation of kidney disease. He was admitted to another hospital via his GP with sepsis. He has no relevant past medical history. His blood results on admission to hospital were: • Serum creatinine 450 µmol/L • Urea 25 mmol/L • Potassium 5.6 mmol/L • Sodium 140 mmol/L • Corrected calcium 2.8 mmol/L • Haemoglobin 10.1 g/L • WCC 15 109/L.

Q1. He has been transferred to your ward. As the ward pharmacist, what further information would you like to know?

Q2. At the previous hospital he was given gentamicin, vancomycin and pamidronate? Could any of these contribute to his renal failure?

Q3. What would be your pharmaceutical care plan at this stage? The 24-hour urine collection shows the presence of Bence-Jones proteins and the medical team plan to biopsy a kidney. The biopsy shows myeloma cast cell nephropathy. The medical team refer the patient to the haemato-oncology team for treatment options. The patient is commenced on haemodialysis and plasma exchange on alternate days. The haemato-onocologist recommends C-Thal-Dex therapy.

Q4. Would your pharmaceutical care plan change at this stage? Outline a new pharmaceutical care plan.

Q5. After 10 days Mr CB remains haemodialysis-dependent as his renal function has not improved. The renal team would like to treat Mr CB’s anaemia and commence an erythro- poesis-stimulating agent. What dose would you recommend? Are there any other factors to consider? 224 Chapter 17 • Myeloma and kidney disease

References steroids in myeloma complicated by renal failure. Br J Cancer 1990; 61: 765–766. 7. Alexanian R, Dimopoulos MA, Delasalle K, 1. Smith A, Wisloff F, Samson D. Guidelines on the Barlogie B. Primary dexamethasone treatment of diagnosis and management of multiple myeloma multiple myeloma. Blood 1992; 80: 887–890. 2005. Br J Haematol 2005; 132: 410–451. 8. Myeloma Trialists’ Collaborative Group. 2. Winearls CG. Acute myeloma kidney. Kidney Int Combination chemotherapy versus melphalan 1995; 48: 1347. plus prednisone as treatment for multiple 3. Blade J, Fernandes-Llama P, Bosch F et al. Renal myeloma: an overview of 6633 patients from 7 failure in multiple myeloma. Presenting features randomised trials. J Clin Oncol 1998; 16: and predictors of outcome in 94 patients from a 3832–3842. single institution. Arch Intern Med 1998; 158: 9. The Nordic Myeloma Study Group. Interferon- 1889. alpha 2b added to melphalan-prednisone for 4. Clark AD, Shetty A, Soutar R. Renal failure and initial and maintenance therapy in multiple multiple myeloma: pathogenesis and treatment myeloma. A randomized, controlled trial. Ann of renal failure and management of underlying Intern Med 1996; 124: 212–222. myeloma. Blood Rev 1999; 13: 79–90. 10. MERIT (Myeloma Renal Impairment Trial). A 5. Pozzi C, Pasquali S, Donini U et al. Prognostic randomized controlled trial of adjunctive plasma factors and effectiveness of treatment in acute exchange in patients with newly diagnosed renal failure due to multiple myeloma: a review multiple myeloma and acute renal failure. Renal of 50 cases. Clin Nephrol 1987; 28: 1–9. Association and the UK Myeloma Forum, 6. Aitchison RG, Reilly IAG, Morgan AG, Russell Protocol 1.6, September 2005. NH. Vincristine, adriamycin and high dose 18

Palliative care

Fliss Murtagh, Emma Murphy and Hayley Wells

Technological and medical advances bring new patient’s death is as gentle and symptom-free as possibilities for prolonging life. In the UK in possible. During the last stages of a patient’s life, 1951 there were 300 people aged 100 and over; medical responsibility shifts from sustaining life by the year 2031, it is estimated that this figure to enabling a person to die comfortably. will reach 36 000.1 The World Health Organiza- Removal of active treatment is not abandon- tion warns that the health impact could be ment of the patient, but rather ensures that enormous, predicting a big rise in the preval- the patient is as comfortable as possible and ence of chronic diseases, including renal disease. has a dignified death, with appropriate family The percentage of the population on dialysis is support. increasing by around 10% per annum and this There is also a group of dialysis patients, increase contains disproportionate numbers of again particularly older people and those with older patients with poor functional status and co-morbidity, who decide to withdraw from extensive co-morbidity.2 dialysis therapy. The mean survival time There is a greater recognition within renal from the last dialysis session has been stated to teams that dialysing these patients may not be about 9 days, although the range extends greatly improve survival but can negatively from 1 to 42 days.6 Advance planning, good impact on quality of life3,4 and growing num- symptom management and high-quality end bers of patients are therefore being managed of life care is also critical for this group of conservatively (choosing, in conjunction with patients. their renal team, not to have dialysis). The In the literature, patients with renal failure National Service Framework (NSF) for Renal who follow a conservative management path- Services requires that people with renal failure way have underreported symptoms, but work at should receive information about the choices Guy’s and King’s College Hospitals has demon- available, including conservative management, strated a high burden of symptoms with over and that, if appropriate, they should have a half of the patients reporting poor mobility, palliative care plan built around their needs.5 weakness, pain and itching and over one-third The NSF also states that people who choose a reporting poor appetite, difficulty sleeping, non-dialysis pathway should continue to drowsiness, dyspnoea and constipation (per- receive all other medical care and that a ‘“no- sonal communication). This study also indi- dialysis” option is not a “no-treatment” option;5 cated that these symptoms became more maximal conservative management including prevalent as renal function worsened. active symptom management can relieve many Established palliative care services are symptoms and substantially improve quality increasingly accepting referrals of patients with of life.’ renal failure but compared to malignant condi- Patients choosing this management option tions the numbers of patients are small. may survive for months or even years, but However, the help of the local specialist palliat- when end of life does approach, healthcare pro- ive care team should always be sought if symp- fessionals have a duty to ensure that each tom control is difficult.

225 226 Chapter 18 • Palliative care

Prescribing for symptom control in to moisten the mouth in fluid-restricted patients with stage 5 chronic kidney patients, using artificial saliva, and, if persistent, disease managed without dialysis pilocarpine 5 mg three times daily can be con- sidered. Relaxation of dietary restrictions may be appropriate with referral to a renal dietician The main principles of symptom management for advice. Addition of zinc 220 mg four times in patients with stage 5 chronic kidney disease daily may be effective to treat taste disturbances. 2 (eGFR <15 mL/min/1.73 m ) being managed If these options fail, direct pharmacological without dialysis are common to other palliative measures can be used to treat anorexia. These care areas, but there are specific issues that arise, include: including the very altered pharmacokinetics and pharmacodynamics when eGFR is less • Dexamethasone 2–4 mg daily for up to four than 15 mL/min/1.73 m2. These patients have weeks. Prolonged use is not recommended as increased sensitivity to drugs, with reduced the effects are relatively short lived. If an renal excretion, accumulation of metabolites, improvement is noted the dose can be decreased protein binding and increased per- weaned down gradually over several weeks; if meability of the blood–brain barrier, all increas- no benefit is noted it can be reduced and ing risk of toxicity. In the latter stages of stopped over a much shorter period of time. treatment the renal function is often unknown • Medroxyprogesterone 100–200 mg daily as blood samples may not be taken, so drugs improves appetite but has an unknown mode need to be initiated at low doses, often with of action, with more prolonged benefit than increased dosing intervals, and then titrated cortico steroids. It is therefore more suitable slowly to effect with the knowledge that the for those with a lengthier prognosis. A 1–2 drug may accumulate very rapidly. week trial is recommended but it is contra- The symptoms most likely to occur are indicated in those patients with a thrombo- anorexia, constipation, dyspnoea, fatigue, embolic risk. nausea and vomiting, pain, pruritus and restless • Thalidomide and omega-3 fatty acids have legs syndrome. It is important to identify the also been suggested, but there is currently cause of any symptoms as far as possible and insufficient supporting evidence. treat the underlying cause if feasible before con- sidering purely symptomatic management. Constipation

Constipation is common in this group of Anorexia patients with multifactorial causes: drugs, poor dietary intake, reduced mobility, dehydration, Anorexia is an inevitable consequence of depression and reduced muscle tone through advanced renal failure and uraemia, and a dis- immobility. tressing symptom for patients and family. Management Management A thorough assessment is required to treat There may be contributing reversible factors to reversible causes of constipation where possible, loss of appetite (e.g. constipation, diarrhoea, with acute management to treat current con- oesphagitis, nausea and vomiting, gastroparesis, stipation and active management to prevent dry mouth, oral candidiasis, dysphagia and further recurrence. Mobility, adequate dietary anxiety), which should be actively treated intake including sufficient fibre and fluid before considering direct pharmacological meas- should be encouraged before laxatives are ures. Dry mouth should be treated by ensuring prescribed. the patient is not dehydrated, if possible stop- If these measures fail, laxatives are required. ping drugs that exacerbate dry mouth, using ice The most commonly prescribed laxatives are Prescribing for symptom control 227

Table 18.1 Commonly prescribed laxatives used to treat constipation in palliative care patients with renal failure

Drug Dose Mode of action Comments

Lactulose 10–20 mL BD/TDS Osmotic – softens by Should be used with a stimulant laxative when retaining fluid in bowel prescribed with opioids Senna 2–4 tablets BD Stimulant Bisacodyl 5–10 mg OD/BD Stimulant Co-danthramer 5–20 mL BD Stimulant and softener Avoid in urinary or faecal incontinence as can cause danthron burns Docusate sodium 100–200 mg BD Stimulant and softener Movicol 1–3 sachets daily Osmotic Fluid restrictions and/or poor oral intake may constrain use

BD, twice daily; OD, once daily; TDS, three times daily.

listed in Table 18.1. If the patient is on opioid diuretics. Table 18.2 lists some of the more com- medication, it is usual to require both a ‘sof- monly used diuretics. tener’ and a stimulant in moderate to high Bronchodilators and other therapy may be doses. useful for symptomatic treatment if there is co- existing respiratory disease and bronchospasm. There are some simple non-drug measures Dyspnoea that can also be used to reduce dyspnoea: • Position – sitting upright rather than lying The most common causes of dyspnoea in increases the vital capacity of the lungs patients with renal impairment are anaemia or • Cool air from a fan or an open window fluid overload (related to renal disease), pul- • Oxygen therapy if hypoxia is confirmed or monary oedema (usually related to co-existing suspected (usually when there is co-existing cardiovascular disease) or co-existing respiratory cardiac or respiratory disease) disease. Active management of the underlying • Reassurance and active anxiety management, cause of the dyspnoea is the most important including work with families. strategy, although as the patient reaches the end of life and dyspnoea is likely to become more Dyspnoea is very commonly associated with problematic, active treatment of any underlying anxiety, often in an escalating cycle (anxiety cause may no longer be feasible, and purely causing worsening dyspnoea, which triggers symptomatic management becomes important. worsening anxiety, and so on). It is important to inform, educate and support the patient and family and to identify psychological and phys- Management ical triggers to episodes. Regular use of relax- Significant symptoms of dyspnoea can be ation techniques (e.g. aromatherapy, massage) caused by anaemia and although any anaemia and cognitive behavioral therapy can help to is likely to be due to renal failure, other causes reduce anxiety and may be effective in reducing should be considered and excluded. If patients dyspnoea. have months to live, anaemia should be actively When treatment of the underlying cause and managed (see Chapter 5). Planning with the other measures can offer no more relief, then patient how they would like to be treated in the pharmacological interventions to control the future is essential if they become acutely symp- symptoms are indicated. Untreated moderate or tomatic, as not all patients will choose to be severe dyspnoea at the end of life is particularly admitted to hospital for aggressive intravenous distressing; no patient should be allowed to 228 Chapter 18 • Palliative care

Table 18.2 Commonly prescribed diuretics used to treat fluid overload in palliative care patients with renal failure

Diuretic Dose Mode of action Comments

Furosemide Oral 80 mg–2 g daily Loop diuretic Furosemide acts within 1 hour of oral (2 g rarely used) administration and diuresis is usually IV 40 mg–1 g daily complete within 6 hours. Doses at the (Note: 500 mg orally = higher end of the range are often used in 250 mg IV) this group of patients Bumetanide Oral 1–5 mg in severe cases Loop diuretic 1 mg of bumetanide = 40 mg furosemide IV 1–2 mg repeated after at low doses (avoid direct substitution at 20 min high doses) Metolazone 5–10 mg increased to Acts synergistically Introduce cautiously – may result in 20 mg daily with loop diuretics profound diuresis. Monitor fluid balance – not used alone carefully and review patient soon after commencement. Monitor for hypokalaemia, dehydration and rising urea

remain dyspnoeic and distressed in their last Nausea and vomiting days of life. The mainstay of pharmacological symptomatic management is benzodiazepines. Nausea and vomiting is common in patients Uraemic patients are more sensitive to the with end stage renal failure, resulting in the cerebral effects of these drugs, and often very inability to take diet or drugs orally, and patients small doses are sufficient. If the patient is still can quickly become dehydrated. Nausea and able to take oral medication, then a shorter vomiting may frequently be multifactorial and acting benzodiazepine such as sublingual specific causes need to be identified. Drugs (e.g. lorazepam 0.5 mg is often effective. If the opioids) or toxins such as high serum urea or cal- patient is too ill for oral medication and in cium may cause nausea, which is usually per- the last few days of life, then midazolam 2.5 mg sistent and not relieved by vomiting. In this subcutaneously is usually effective. The dose situation retching may feature more promi- should be repeated as required, but is rarely nently than actual vomiting. Gastric motility needed more than every 4–6 hours, and some- problems (especially common in diabetic times much less frequently. patients) usually cause post-prandial vomiting of Opioid medications are used for breathless- undigested food, which often relieves the nausea ness in patients with cardiac and respiratory and may be accompanied by bloating, epigastric dyspnoea – in much lower doses than for pain, fullness, flatulence, hiccough or heartburn. but with supporting evidence.7 There is no Nausea and vomiting associated with gastritis is supporting evidence for the use of opioids for often associated with heartburn or epigastric dyspnoea in renal patients, and there are many pain, which is sometimes worse after eating. constraints on the use of opioids in patients Emesis can also be associated with movement or with eGFR less than 15 mL/min who are not constipation. dialysed (see Chapter 15), but nevertheless, If nausea is established and vomiting fre- for a dyspnoeic renal patient near the end of quent or severe, then oral drugs will not be life and who perhaps has co-morbid cardiac absorbed and a continuous subcutaneous infu- or respiratory disease, they may be very use- sion is indicated. Once symptoms have settled ful. Starting doses are usually half those for (but not before) subcutaneous antiemetics can pain. then be replaced by oral antiemetics. Prescribing for symptom control 229

Management should be treated actively as pain may some- times not improve until this is addressed. The aim of treatment is to identify and treat the The principles of the World Health Organ- suspected cause as shown in Table 18.3. ization analgesic ladder should be followed for primary analgesia as in other palliative care patients, and constant or frequent pain should be Pain treated with regular, not ‘as required’ analgesia.10 Initially step 1 analgesia (e.g. paracetamol) Pain is underrecognised and undertreated in should be started, unless pain is severe. The next this group of patients.8,9 It is hard to manage step (step 2 opioids for moderate pain) should be due to the constraints reduced renal function proceeded to if the analgesic from the current puts on the use of medicines. The little evidence step has not controlled the pain adequately. Step that is available on pain in this group of 3 opioids for severe pain should be used if step 2 patients suggests the pain is often due to co- medication is not enough. At each step, another morbidities rather the impaired renal function.8 drug within the same step should not be tried if Examples include neuropathic pain in, for pain is inadequately controlled at that step. example, diabetic neuropathy, and bone pain Using opioids when eGFR is less than 15 mL/ from osteoporosis, but these pains still need min/1.73 m2 is challenging, and can be complex, active management. Anxiety or depression but it is important not to leave pain untreated

Table 18.3 Commonly prescribed antiemetics used to treat nausea and vomiting in palliative care patients with renal failure

Suspected cause Drug of choice Dose Comments

Drug-induced or uraemia (First Haloperidol 0.5–2 mg daily Sedating, accumulation and step is to stop causative drugs if increased cerebral sensitivity in possible. Opioid-induced nausea renal failure usually settles spontaneously after about 7–14 days of treatment) Gastric motility problems Metoclopramide 5–10 mg TDS Do not use in bowel obstruction with colic. Do not use with cyclizine Domperidone 10–20 mg TDS Rectal route available Gastritis Lansoprazole 30 mg OD Omeprazole 20 mg OD Ranitidine 150 mg BD Second line (if severe or unknown (Consider asking or multifactorial causes) advice of specialist palliative care team at this stage) Levomepromazine Starting dose: Very sedating at higher doses may be appropriate a3 mg–6.25 mg but needs cautious OD orally or use in low doses 2.5 mg because of increased subcutaneously cerebral sensitivity

BD, twice daily; OD, once daily; TDS, three times daily. a 6 mg tablets available on named-patient basis. 230 Chapter 18 • Palliative care because of this. The analgesics discussed are out- have a high incidence of drowsiness and lined in Table 18.4. lethargy but it can be a very useful step 2 anal- Adjuvant medicines, such as non-steroidal gesic in this group of patients. anti-inflammatory drugs (NSAIDs) (for musculo- skeletal or soft tissue pain), antidepressant or Step 3 analgesia anticonvulsant medication (for nerve pain), are important considerations. NSAIDs can be used if The choice of step 3 analgesic will depend on the benefits of analgesia outweigh the risk of the severity and acuteness of the pain, whether potential further reduction in renal function. If the patient is in primary or secondary care, NSAIDs are used, a gastro-protective agent (e.g. appropriate route of administration, and avail- omeprazole) should be co-prescribed. ability of drugs and preparations. If the patient has severe pain and is in hospi- tal, then the subcutaneous route is preferable to Step 1 analgesia gain rapid control of the pain. Once severe pain Paracetamol (maximum dose 1 g three times is controlled by the subcutaneous route, anal- daily if eGFR less than 15 mL/min/1.73 m2). gesia can either be switched to the trans-dermal routes or oral routes, or (if the patient is unable to take oral drugs) continued in a continuous Step 2 analgesia subcutaneous infusion via syringe driver.

Codeine and dihydrocodeine Buprenorphine Codeine and dihydrocodeine are hepatically Buprenorphine acts as a potent partial mu- metabolised with the majority of the metabolites opioid receptor antagonist, kappa-opioid re- pharmacologically active and renally excreted. In ceptor antagonist and a weak delta-opioid end stage renal failure this renal clearance is receptor agonist. The parent drug is excreted via reduced and there have been reports of serious the biliary system, but the metabolites side-effects, including severe hypotension and buprenorphine-3-glucuronide and norbupre- respiratory arrest.11–13 If possible, both these norphine, are excreted renally.18 There has been analgesics should be avoided in patients with an little published pharmacokinetic data in eGFR less than 15 mL/min/1.73 m2 who are not patients with renal impairment so it cannot be dialysed, but this may not always be practical. If wholly recommended. The effects and possible used, the dose should be reduced to 25–50% of toxicity of the metabolites is unclear. However, normal with careful monitoring of blood pressure the partial biliary excretion may make it more and respiratory rate. Constipation should be appropriate for use in renal impairment; if used, anticipated and therefore laxatives co-prescribed. it should be prescribed cautiously, with dose reduction, increased dosing interval, and careful Tramadol monitoring, pending further evidence. Tramadol acts peripherally and centrally on mu- opioid receptors and by inhibiting monoamine Fentanyl and alfentanil (noradrenaline (norepinephrine) and serotonin) Fentanyl is a potent synthetic opioid that is reuptake. Ninety per cent of the drug is excreted hepatically metabolised to norfentanyl and renally and there is a twofold increase in the other inactive metabolites with less than 10% elimination half-life in end stage renal failure.14 of the parent drug excreted renally.14,16 When In this group of patients both the dose should single bolus doses are given to patients with be reduced and the dosing interval increased, to renal impairment no dose modifications appear a maximum of 50 mg every 12 hours.15 to be required.19 However, there are few data on Tramadol may also lower fit threshold in this the pharmacokinetics when the drug is given group of patients with a high urea so use in repeatedly or by infusion. Studies have shown those at risk of convulsions for other reasons that fentanyl may accumulate with extended should be avoided. Clinically, tramadol may administration and that there is considerable Prescribing for symptom control 231

Table 18.4 Analgesics used to treat pain in palliative care patients with eGFR <15 mL/min and without dialysis

Drug Dose Route Frequency Comments

Step 1 Paracetamol 1 g PO TDS Step 2 Codeine Not recommended Dihydrocodeine Not recommended Tramadol 50 mg Orally Every 12 hours Use with caution Step 3 Buprenorphine 200–400 µg Sublingual Every 8 hours Safer than morphine or diamorphine ‘5’ patch upwards Transdermal every 7 days but limited evidence so use with ‘35’ patch upwards Transdermal every 72–96 caution. Greater incidence of nausea hours than other step 3 opioids, so consider avoiding if patient already nauseous. Theoretically, ‘ceiling’ effect, and both agonist and antagonist properties, so perhaps avoid if pain is escalating, or alternative step 3 opioid likely to be used later Fentanyl Starting dose Subcutaneous 3–6 hourly No appropriate oral preparation 25µg as according to available for chronic pain. (Oral required need transmucosal fentanyl is only suitable for breakthrough or incident pain.) ‘25’ patch upwards Transdermal every 72 hours The patch is effective once pain is stable on other opioid, but is not advisable if the patient is opioid naïve Alfentanil Use in continuous SC infusion, when volume makes larger doses of fentanyl impractical Hydromorphone: Limited evidence of safety, but like As Palladone 1.3 mg upwards Oral Every 6–8 oxycodone, may be used by some hours practitioners despite limited evidence As Palladone SR 2 mg upwards Oral Every 12 hours of safety. Monitor for accumulation with long term use Methadone Reduce dose by Safe when eGFR <15 but needs 50–75% specialist palliative care or pain team advice on prescribing, as wide inter-individual variations and risk of (late) accumulation Morphine Starting dose Subcutaneous 4–12 hourly Not recommended (but see text re. 2.5 mg as required short-term use) Diamorphine Starting dose Subcutaneous 4–12 hourly Not recommended (but see text re. 1.25 mg as required short-term use) Oxycodone: Used but limited evidence of safety. as OxyNorm 2.5 mg upwards Oral Every 8–12 No upper dose limit, provided it is hours titrated slowly, is effective for the pain, as OxyContin 5 mg upwards Oral Every 12 hours and there are no adverse effects. Start with OxyNorm, to gain rapid control of the pain, and consider switch to OxyContin later for ease of administration (but beware accumulation with longer term use) 232 Chapter 18 • Palliative care inter-patient variability.16 Alfentanil has a much is also an important route of elimination. There shorter half-life than fentanyl, but similarly has is a large inter-patient pharmacodynamic and inactive metabolites, and, with fentanyl, per- pharmacokinetic variation with methadone treat- haps the best evidence for safety in severe renal ment, and it accumulates on repeated adminis- impairment.14,16 tration due to its high volume of distribution. For the management of moderate or severe There is evidence in two patients with chronic chronic pain in advanced renal impairment, renal failure that plasma concentrations were no fentanyl or alfentanil are therefore probably the higher than in those with normal renal function, optimal step 3 opioids. In order to prevent accu- suggesting that faecal excretion may compensate mulation, the dose should be reduced (e.g. 75% for impaired renal excretion.25 For this reason, of usual dose when eGFR is between 15 and methadone may be a good choice of opioid in 50 mL/min/1.73 m2 and 50% dose when eGFR severe renal impairment, but because of the prob- is less than 15 mL/min/1.73 m2), with careful lems with inter-patient variability and accumula- and frequent monitoring for adverse effects. tion, it is advisable that it is only prescribed by Alfentanil, however, has the disadvantage of a specialists familiar with its use and monitoring. short half-life (1–2 hours). This makes it in- Dose reduction of 50–75% and close monitoring appropriate for prn (as required) use – pain will is advised.15 recur quickly and additional doses may need to be given frequently. For this reason, fentanyl is Morphine and diamorphine a better choice. At the end of life, however, if Morphine is metabolised hepatically to renally large doses of fentanyl are required in a continu- excreted metabolites, morphine-3-glucuronide ous subcutaneous infusion (after careful titra- (M3G) and morphine-6-glucuronide (M6G), tion of analgesic requirements against the pain), which accumulate in significant renal impair- then it is sometimes necessary to switch to ment.14,16 The metabolite M6G may be more alfentanil, because it is available in more con- potent than morphine and depresses the central centrated preparations. nervous system. M6G also crosses the blood– brain barrier slowly and once there its action is Hydromorphone prolonged. M3G appears to have little analgesic Hydromorphone is a morphine analogue with activity but may antagonise the effect of M6G. a short duration of action and is hepatically Diamorphine is rapidly deacetylated to mor- metabolised to its main metabolite hydro- phine and M6G and acts similarly to morphine. morphone-3-glucuronide (H3G). This is cleared Because alternative strong opioids (such as renally and accumulates in renal failure.20,21 fentanyl) are now available, morphine and There is evidence that H3G is neuroexcitatory diamorphine are no longer first-choice opioids in both animals22 and humans.23 There are when eGFR is less than 15 mL/min and patients concerns about the use of hydromorphone in are not dialysed. In the last few days of life, this group of patients as a single-dose study indi- morphine and diamorphine use can increase cated accumulation of the drug and its meta- the risk of terminal agitation due to accumula- bolite in renal failure.20 Other reports in mild tion of metabolites.17 They should therefore renal failure suggested that the drug was better only be used where there is moderate or severe tolerated.24 However, the evidence for hydro- pain and alternative opioids are unavailable (e.g. morphone in renal failure is limited, and dose in the community setting), and in the short reduction, titration and monitoring is essential. It term (e.g. for one or two doses). Efforts should may be preferable to morphine or diamorphine, be made to try and anticipate the requirements but (like oxycodone) cannot be recommended in for a step 3 opioid before it occurs, and to pro- the absence of evidence of safety. vide the most appropriate drug (e.g. fentanyl) in case of need. This requires anticipation and Methadone planning, ensuring that patients nearing death Methadone is hepatically metabolised and the at home have prn medication provided before metabolites are predominantly faecally excreted, need, so that district nurses and GPs can then although renal excretion of the unchanged drug administer when required. If morphine or Prescribing in the last few days of life 233 diamorphine is used, the initial dose should be liberal emollients (e.g. aqueous cream). Nails small (e.g. 2.5 mg SC morphine) and titrated to should also be kept short and patients advised response. If a continuous SC infusion is to keep cool with light clothing, tepid baths or required, morphine or diamorphine are not the showers. There is limited evidence for drug opioids of choice as they will be excreted slowly, therapy with no one preparation recom- if at all; any CNS effects will be prolonged. mended.27 However, individual patients do report significant benefit, with some of treat- Oxycodone ments outlined in Table 18.5. Time should be Oxycodone is metabolised hepatically to the taken to discuss with the patient the need to active metabolites noroxycodone and oxymor- persist with any one drug, and to explaining phone, with less than 10% of the parent drug how to minimise side-effects where possible. excreted in the urine. A study of 10 uraemic patients demonstrated a reduced elimination in renal failure patients with a large inter-patient Restless legs syndrome variation.26 There is currently insufficient evid- ence to confirm the safety of oxycodone in There are limited studies on uraemic restless legs renal failure patients and if used it should be syndrome, so much evidence is extrapolated monitored carefully with reduced doses of from management of idiopathic restless legs. In 75% of normal dose when eGFR is 10–50 mL/ uraemia, restless legs are thought to be associ- min/1.73 m2, and 50% of normal dose when ated with anaemia, iron deficiency and hyper- eGFR is less than 10 mL/min/1.73 m2.15 Like phosphataemia.28,29 Psychological factors may hydromorphone, it is a better choice than also play a role. morphine or diamorphine. Management Pruritus Treatment should involve the reduction of potential exacerbating drugs (tricyclic anti- Pruritus is a difficult symptom to manage, and depressants, selective serotonin uptake inhib- advice should be sought early from a specialist itors, lithium and dopamine antagonists) and palliative care team if the first options are not correction of anaemia, iron deficiency and successful. Although uraemia is the most likely hyperphosphataemia. Drugs used to treat rest- cause, other common causes of pruritus need less leg syndrome are outlined in Table 18.6. to be considered if the symptoms are not resolv- ing (e.g. other skin disorders, scabies, liver impairment). Prescribing in the last few days of life

Management When managing a patient in the last few days Management of renal failure should be optim- of life, care is modified to focus on making the ised and for certain patients, high serum phos- patient as comfortable as possible with stopping phate levels may contribute to pruritus, so a of all unnecessary blood tests, monitoring and referral to a renal dietician for dietary advice, non-essential medicines. At the end of life, not and prescription of phosphate binders to reduce only do the symptoms discussed previously phosphate levels should be considered. Hyper- continue to require management but other parathyroidism may also be a contributory symptoms also require treatment, such as rest- factor so parathyroid hormone should be lessness/agitation, or retained respiratory secre- monitored and treated with alfacalcidol as tions. Midazolam subcutaneously is effective for appropriate, while monitoring for hyper- restlessness and agitation but patients with calcaemia. severe renal impairment are much more sensit- Dry skin both causes and contributes to pru- ive to the cerebral effects of all benzodiazepines ritus, and should be treated very actively using – doses should be reduced and monitored 234 Chapter 18 • Palliative care

Table 18.5 Commonly prescribed drugs used to treat pruritus in palliative care patients with renal failure

Drug Dose Comments

Emollients Topical, ad lib Long standing use, but limited evidence of benefit. May be important for those with dry skin Antihistamines Chlorphenamine 4 mg QDS (TDS if Some patients benefit from a sedative effect for nocturnal eGFR <15 mL/min) pruritus – if this is the case, use a sedating antihistamine such as chlorphenamine Cetirizine 10 mg OD (5 mg if eGFR <15 mL/min) Immune modulators Thalidomide 100 mg at night There is evidence that thalidomide may give notable benefit, especially in more severe pruritus. The drug is unlicensed and caution is required to ensure no pregnant or potentially pregnant woman are exposed to the drug (even the handling of it). There is also risk of (reversible) neuropathy Ondansetron 2–8 mg BD A few very small trials have been undertaken with conflicting results. If used, it is also helpful for nausea, but note it is very constipating (laxative should be co-prescribed). Capsaicin 0.025% cream Impractical unless pruritus is localised. If used, local burning applied four times daily may occur, and deter continuation Gabapentin After dialysis Not advisable for use in the non-dialysed patient with eGFR <15 mL/min due to near 100% renal clearance and rapid accumulation

OD = once daily, BD = twice daily, TDS = three times daily, QDS = four times daily.

Table 18.6 Commonly prescribed drugs used to treat restless legs syndrome in palliative care patients with renal failure

Drug Starting dose Comments

Co-careldopa 12.5/50 at night Restless legs syndrome may become, with time, worse in about 80% of cases. This correlates with greater accumulated dose of levodopa, so the lowest dose for shortest duration should be used. Pulsed dosing can be considered Pergolide 25 µg OD Nausea is a common side-effect Clonazepam 250–500 µg Very limited evidence of benefit. Can cause day-time drowsiness and cognitive impairment Opioids Extremely limited evidence of benefit, but may be appropriate to consider if there is concurrent pain. Follow the guidelines in the pain section as to opioid selection and dosing

OD = once daily. carefully. Often 2.5 mg 6–8 hourly is sufficient. hyoscine butylbromide 20 mg as required or For retained respiratory secretions it is advisable glycopyrronium 0.2 mg as required. to avoid hyoscine hydrobromide (because of Every patient should have an analgesic, possible cerebral effects), and use either antiemetic, sedative and medication for retained Conclusions 235 secretions available for prn use in the last few Specialist palliative care services see very few of days of life, whether or not they have required these patients and this is a newly recognised these medications before. This is important, area for renal teams. Pharmacists, as part of especially if patients are discharged from hospi- this team can have a great influence on improv- tal to die at home, and hospital pharmacists will ing management of patient symptoms and need to facilitate this anticipatory prescribing of ensuring a patient’s end of life is as comfortable additional medications to take home. as possible.

Acknowledgements Conclusions We would like to thank Mee-Onn Chai, renal For patients following a conservative pathway pharmacist at King’s College Hospital, for her or withdrawing from dialysis, symptom man- help with this chapter. agement and prescribing can be challenging.

C CASE STUDY

Mr RP is a 63-year-old man with a history of end stage renal failure secondary to diabetic nephropathy. Complications of IDDM include nephropathy, retinopathy and neuropathy. He was diagnosed with metastatic prostate carcinoma in late 2005. His PSA was over 900 on diagnosis, then dropped but has been rising again despite goserelin (Zoladex). Further hormone manipula- tion did not improve disease control. Subsequent investigations with CT and bone scan showed him to have bone metastases. He has been informed by the urology team that he has a poor prognosis and a likely life expectancy of 1 year. He has opted for conservative management of his renal failure. He has no follow-up outpatient appointment with oncology. He was keen to attend the renal palliative clinic to discuss symptoms and the future. Biochemistry review reveals serum creatinine 430 µmol/L, urea 25 mmol/L and eGFR 13 mL/min/1.73 m2. His current medications are: • Lansoprazole 30 mg once daily • Gliclazide 40 mg once daily • Alfacalcidol 0.5 µg once daily • NeoRecormon 5000 units weekly • Simvastatin 10 mg every night • Ramipril 2.5 mg once daily • Adcal 1 three times daily • Lactulose 10 mL twice daily • Senna 2 tabs at night • Docusate sodium 2 tablets twice daily • Co-danthramer 10 mL twice daily • Zoladex every 3 months • Co-codamol 2 tablets four times daily. (continued overleaf) 236 Chapter 18 • Palliative care

C CASE STUDY (continued)

When seen in clinic his main complaint is of pain. He has severe pain related to his left lower limb. He complains of aching over his anterolateral thigh and lateral leg. This pain is graded 8 out of 10 at its worst. It is exacerbated by sitting for long periods of time, walking and by try- ing to extend his hip or knee. He also complains of severe pain related to his right shoulder, which is aching in nature and radiates all the way down his right arm to his wrist. He grades this pain 7 out of 10 and it is exacerbated by lying down. At present he is taking co-codamol maximum dose particularly at night for pain. He finds this has very little impact on the pain and reduces it by approximately 20% at the most. He tolerates it fine.

Q1. What are the possible causes of his pain? In addition to his pain he complains of nausea and vomiting. This is not related to his constipa- tion and there is no associated colic. He becomes increasingly nauseous after eating and can only manage a few mouthfuls before feeling full. His vomiting is occasional, consists of undigested food, and relieves his nausea. This pattern of nausea and vomiting is suggestive of gastric stasis which may be related to his diabetes. He also complains of constipation. He can go 7–10 days without opening his bowels and then alternates to diarrhoea due to an increase in his laxatives.

Q2. How would you manage his nausea and vomiting? He sleeps poorly largely due to his pain. His sleep is particularly interrupted by a stabbing pain in his left forefoot which occurs at night but is not helped by sitting out and lowering his leg. He is known to have calcified vessels on recent Doppler scans.

Q3. What might be causing his pain? What other treatment would you consider to relieve his pain? He lives with his wife at home. He manages reasonably well and his wife is clearly very involved in his care. Due to his visual impairment his wife organises his dosette box on a weekly basis. He can manage to go up and down the stairs at home but this wears him out. He does not have a bathroom downstairs at home, which can present some difficulties. His wife works full time. They have two children who do not live at home. He is reviewed one month later in the renal palliative clinic and his main concern is his low mood. He feels quite hopeless and negative and gains no pleasure from anything. He acutely feels the loss of his physical fitness and his former role. He also suffers somatic symptoms in the shape of early waking and poor appetite and lethargy. He acknowledges he is depressed. He does not have a past history of treatment for this. On further questioning he describes suicidal ideation and intent. He requests that this is not discussed with his GP. When reviewed in clinic two months later he has clearly deteriorated. He finds the transport issues particularly difficult due to his poor mobility and finds this has significant impact on his quality of life. His general level of functioning is now very poor. He is essentially wheelchair- or bed-bound. He has had several falls and recent fracture of his right humerus. He feels much more vulnerable and unsafe at home. His wife is out at work during the day and so he spends long periods of time on his own. He is keen to know if he has any rehabilitation potential with his legs and mobility.

Q4. What would you discuss at this stage? What medications could you ensure were prescribed for the last few days of life? References 237

References medications – dosage guidelines. Prog Palliative Care 2003; 11: 183–190. 16. Davies G, Kingswood C, Street M. Pharmaco- 1. Government Actuary’s Office. National Population kinetics of opioids in renal dysfunction. Clin Projections. Series PP2 No 20. London: Office for Pharmacokinet 1996; 31: 410–422. National Statistics, 1996. 17. Kirkham SR, Pugh R. Opioid analgesia in uraemic 2. Ansell D, Feest T, Rao R et al. UK Renal Registry patients. Lancet 1995; 345: 1185. Report 2005. Bristol: UK Renal Registry, 2006. 18. Hand CW, Sear JW, Uppington J et al. 3. Munshi SK, Vijayakumar N, Taub NA et al. Buprenorphine disposition in patients with renal Outcome of renal replacement therapy in the impairment: single and continuous dosing, with very elderly. Nephrol Dial Transplant 2001; 16: special reference to metabolites. Br J Anaesth 128–133. 1990; 64: 276–282. 4. Smith C, Silva-Gane M, Chandna S et al. 19. Coral IM, Moore AR, Strunin L. Plasma concen- Choosing not to dialyse: evaluation of planned trations of fentanyl in normal surgical patients non-dialytic management in a cohort of patients with severe renal failure. Br J Anaesth 1980; 52: with end-stage renal failure. Nephron Clin Pract 101P. 2003; 95: c40–c46. 20. Durnin C, Hind ID, Wickens MM et al. 5. Department of Health. National Service Framework Pharmacokinetics of oral immediate-release for Renal Services – Part 2 Chronic Kidney Disease, hydromorphone in subjects with renal impair- Acute Renal Failure, and End of Life Care. London: ment. Proc West Pharmacol Soc 2001; 44: 81–82. Department of Health, February 2005. 21. Babul N, Darke AC, Hagen N. Hydromorphone 6. Cohen LM, McCue JD, Germain M, Kjellstrand metabolite accumulation in renal failure. J Pain CM. Dialysis discontinuation. A ‘good’ death? Symptom Manage 1995; 10: 184–186. Arch Intern Med 1995; 155: 42–47. 22. Wright AW, Mather LE, Smith MT. 7. Jennings AL, Davies AN, Higgins JP et al. A sys- Hydromorphone-3-glucuronide: a more potent tematic review of the use of opioids in the man- neuro-excitant than its structural analogue, agement of dyspnoea. Thorax 2002; 57: 939–944. morphine-3-glucuronide. Life Sci 2001; 69: 8. Davison SN. Pain in hemodialysis patients: prev- 409–420. alence, cause, severity, and management. Am J 23. Fainsinger R, Schoeller T, Boiskin M, Bruera E. Kidney Dis 2003; 42: 1239–1247. Palliative care round: cognitive failure and coma 9. Andreucci VE, Fissell RB, Bragg-Gresham JL et al. after renal failure in a patient receiving captopril Dialysis Outcomes and Practice Patterns Study and hydromorphone. J Palliat Care 1993; 9: (DOPPS) – data on medications in hemodialysis 53–55. patients. Am J Kidney Dis 2004; 44 (Suppl 2): 24. Lee MA, Leng ME, Tiernan EJ. Retrospective study S61–S67. of the use of hydromorphone in palliative care 10. Launay-Vacher V, Karie S, Fau JB et al. Treatment patients with normal and abnormal urea and of pain in patients with renal insufficiency: The creatinine. Palliat Med 2001; 15: 26–34. World Health Organization three-step ladder 25. Kreek MJ, Schecter AJ, Gutjahr CL, Hecht M. adapted. J Pain 2005; 6: 137–148. Methadone use in patients with chronic renal 11. Parke TJ, Nandi PR, Bird KJ, Jewkes DA. Profound disease. Drug Alcohol Depend 1980; 5: 197–205. hypotension following intravenous codeine 26. Kirvela M, Lindgren L, Seppala T, Olkkola KT. The phosphate. Three case reports and some recom- pharmacokinetics of oxycodone in uremic mendations. Anaesthesia 1992; 47: 852–854. patients undergoing renal transplantation. J Clin 12. Talbott GA, Lynn AM, Levy FH, Zelikovic I. Anesth 1996; 8: 13–18. Respiratory arrest precipitated by codeine in a 27. Lugon JR. Uremic pruritus: a review. Hemodial Int child with chronic renal failure. Clin Pediatr 2005; 9: 180–188. (Phila) 1997; 36: 171–173. 28. Dinwiddie LC. Restless legs syndrome: not just a 13. Matzke GR, Chan GL, Abraham PA. Codeine problem for dialysis patients. ANNA J 1997; 24: dosage in renal failure. Clin Pharm 1986; 5: 15–16. 655–662. 14. Mercadante S, Arcuri E. Opioids and renal func- 29. Wetter TC. Restless legs syndrome: a review for tion. J Pain 2004; 5: 2–19. the renal care professionals. EDTNA-ERCA J 2001; 15. Broadbent A, Khor K, Heaney A. Palliation and 27: 42–46. chronic renal failure: Opioid and other palliative

19

Dietary management of kidney disease

Diane Green

Dietary treatment is one of the cornerstones of (NIH) in the USA initiated a large multicentre treatment for patients with end stage renal dis- study – the Modification of Diet in Renal ease. In the 1960s many patients were treated Disease (MDRD) study – to investigate the effect with diet alone, using very low-protein diets. of protein restriction on the progression of kid- The renal diet was adapted to dialysis and trans- ney failure. It used four different levels of plantation when renal replacement therapy protein diets ranging from VLPD supplement became available in the 1970s and 1980s. Today, with keto acids and phosphorus restrictions to protein–energy malnutrition, hyperphos- normal intakes. The results were published in phataemia, chronic fluid overload and obesity 1994.2 The results of the study initially showed are the most common challenges, leading to a slower rate in decline of kidney function in long-term complications and adversely affecting the absence of severe proteinuria and hyper- the outcome of treatment. tension, but there appeared to be no further This chapter covers the important issues of advantage to using VLPD.2 dietary management and the role diet plays in As a result of these findings, treatments the treatment of patients with end stage renal became focused on the prevention of complica- failure (ESRF). tions due to uraemia, improving blood pressure control, prevention of malnutrition and dietary phosphorus restriction to prevent renal bone disease. In the absence of malnutrition, healthy Historic pre-dialysis dietary eating principles are also discussed to help intervention reduce the incidence of cardiovascular disease. Research continues into the effects of protein Historically, the dietary treatment for patients restriction on reducing symptoms of uraemia with ESRF was a very low-protein diet (VLPD). and the progression of kidney failure. The best known was the Giovanetti diet which Protein restrictions combined with a very contained 20 g of high biological protein to high calorie diet are again becoming a treat- cover the essential amino acid requirements. It ment option for patients opting not to have was also necessary to ensure the diet provided dialysis but to follow a conservative medical 50 kcal/kg to prevent loss of lean body mass and management route. to maintain nitrogen balance.1 This was com- monly prescribed to patients with ESRF and was the only treatment available to patients until Pre-dialysis dietary management regular haemodialysis became available. In the 1980s there was a renewed interest in low-protein, high-energy diets as partially The nutritional content of a diet for a patient nephrectomised rats showed that protein with chronic kidney diseases should be spe- restriction delayed the progression of renal dis- cifically adapted to their individual needs and ease. In 1985 the National Institutes of Health personal circumstances. Dietary management of

239 240 Chapter 19 • Dietary management of kidney disease kidney disease is very patient-specific. The diet obtained from high biological value proteins, prescribed will depend upon: such as meat, fish, eggs and milk. Once the pro- tein in the diet is reduced, the serum urea and • Stage of chronic kidney disease creatinine levels often decrease. Some patients • Biochemistry notice an improvement in well-being; others • Medical history – diabetes, hypercholesterol- notice a change in taste acuity. emia A daily protein intake of 0.8–1.0 g/kg IBW is • Medical treatment plan recommended,4 with at least 60% from high • Nutritional status biological value protein sources. • The patient’s normal dietary intake. The diet needs to provide sufficient flexibil- Energy ity to enable the patient to continue to lead a normal life and to aid compliance of the dietary The energy values of the different food types are restrictions. The main dietary components of as follows: the renal diet are: protein, energy (fat and car- • Fat 1 g = 9 kcal bohydrate), phosphorus, potassium, sodium, • Carbohydrate 1 g = 4 kcal fluid and other minerals, such as calcium and • Protein 1 g = 3.8 kcal. iron. Patients may be advised to follow none, some or all of the aspects of the diet. The amount of energy required in the diet is as important as the amount of protein. Inadequate energy will lead to protein cat- Protein abolism, weight loss and protein–energy malnu- Protein is an important nutrient in growth and trition. Energy requirements will depend on the repair and maintenance of tissue. activity levels and body size, but a daily intake In the pre-dialysis diet the level of restriction of 35 kcal/kg will be adequate for most patients’ should maintain nitrogen balance and nutri- needs.5 tional status. The amount of protein prescribed To achieve a satisfactory energy intake with is dependent on the patient’s ideal body weight patients on a protein restriction diet, high (IBW) and taking the patient’s residual kidney intakes of fat and/or sugar are often recom- function into consideration (Table 19.1). The mended. This often includes fried food, addi- average daily consumption of protein in the UK tion of cream, high-fat snacks, such as cakes and is 88 g for men and 64 g for women,3 which biscuits, and sugary fizzy drinks to be included means that most patients are advised to reduce in a patient’s diet. their intake of protein foods in their diet. To Obesity should be treated in the early stages ensure that essential amino acids are available of renal failure with a low-calorie diet as this in the diet about 60% of protein needs to can help control a patient’s blood pressure, but

Table 19.1 Protein and energy requirements for pre-dialysis patients

GFR (mL/min) Protein (g/kg IBW) Energy (kcal/kg IBW)

>30 Normal 30–35 20–30 0.6–1.0 (EDTNA/ERCA) 30–35 <19 (not on dialysis) 0.6–1.0 (EDTNA/ERCA) 30–35

GFR, glomerular filtration rate; IBW, ideal body weight; EDTNA/ERCA, European Dialysis and Transplant Nurses Association/European Renal Care Association. From ref. 9. Pre-dialysis dietary management 241 this needs to be carefully monitored as obesity • Milk and milk products (13%) can often mask malnutrition. • Cereal products (13%) • Meat and meat products (15%) Phosphorus (1 mmol = 31 mg P) • Fruit (5%). Phosphate is a major constituent of plants and Progressive renal failure is often complicated animals and so is present in most foods. by hyperkalaemia and may occur when renal Inorganic phosphorus is added to processed function has declined to a glomerular filtration foods, particularly baked goods and carbonated rate (GFR) of 5 mL/min with normal urine drinks, and these provide about 10% of total output. phosphorus intake. The main sources of phos- A dietary intake of no more than 60– phorus in the UK diet3 are: 70 mmol/day (1 mmol/kg IBW) is sufficient to prevent or treat hyperkalaemia. However an • Milk and milk products (24%) anuric non-dialysed patient on a diet contain- • Cereal products (23%) ing 50 mmol K could raise serum potassium by • Meat and meat products (21%) 1 mmol/day despite gastrointestinal adaptation • Vegetables and potatoes (10%). to eliminate dietary potassium in chronic renal Approximately 60% dietary phosphorus is failure (CRF).8 absorbed. A low potassium diet should only be initiated Hyperphosphataemia contributes to the either to prevent or treat hyperkalaemia as well development of renal bone disease through as correcting other possible causes which may the stimulation of parathyroid production.6 have contributed to the rising serum potassium Dietary phosphorus restriction is an essential levels. Some examples of non-dietary causes for part of controlling serum phosphate levels. hyperkalaemia are: However, the need to provide both adequate • Drugs such as angiotensin-converting dietary protein and a palatable diet limits the enzyme (ACE) inhibitors, potassium-sparing degree to which dietary phosphate can be diuretics, non-steroidal anti-inflammatory restricted.7 The appropriate prescription of drugs (NSAIDs) phosphate binders is equally important in con- • Constipation (avoid drugs that contribute to trolling phosphate. constipation, i.e. phosphate binders) All foods with high protein content also con- • Metabolic acidosis tain a fair amount of phosphorus. However, • Increased catabolism some of these foods are an essential part of the • Endocrine abnormalities diet and cannot be eliminated. Table 19.2 lists • Excessive exercise, heat stroke and rhabdo- examples of foods that can be excluded from myolysis, causing hyperkalaemia due to tis- the diet without altering the nutritional quality sue cell destruction.8 of the diet. Most foods contain potassium, but the Potassium (1 mmol = 39 mg K) majority is found in vegetables and fruit. Some varieties contain more than others: basic fruits Potassium is predominantly an intracellular such apples or pears contain 4 mmol of potas- cation and plays a fundamental role in acid– sium per portion, whereas a banana contains base regulation, fluid balance, muscle con- 8 mmol potassium. Staple foods such as pota- traction and nerve conduction. Ninety-five per toes, sweet potatoes and yam, plantain and cent of the body’s potassium in found within green bananas are also high in potassium but the cells. should be included in a potassium-reduced diet. In the average UK diet,3 potassium is derived Cutting up vegetables and potatoes into small from: pieces and boiling in large volumes of water will • Vegetables and potatoes (28%) significantly reduce the potassium content of • Beverages, particularly coffee (15%) those foods (Table 19.3). 242 Chapter 19 • Dietary management of kidney disease

Table 19.2 Low phosphorus diet sheet

Foods with a high phosphorus content Foods with a low phosphorus content

Cereals Bran, All-Bran, Bran Flakes, cereals containing nuts, rye All other breakfast cereals, porridge, puri, chapatti, bread, oatcakes, scones nan, pitta bread, pasta, rice, noodles Dairy products Milk, yoghurt, evaporated and condensed milk, milk Cream, crème fraiche, fromage frais, quark, cottage powder, , hard cheese, cheese spread, eggs cheese and curd cheese, full fat or reduced fat cream cheese such as Philadelphia, roule, Boursin, mascarpone, ricotta, egg white, meringue Meat and meat products Liver, kidney, liver pate and liver sausage, black Beef, veal, lamb, pork, chicken, turkey, sausages pudding Fish and fish products Fish with edible bones such as anchovies, herring, Fresh or smoked fish such as cod, haddock, salmon kippers, sprats, whitebait, fish roe, fish paste Vegetables Pulses such as dried peas, dried beans, lentils, baked All other vegetables beans, chick peas Cakes and biscuits Any cake or biscuit containing chocolate, nuts or All other types of cakes, biscuits, pastries, doughnuts, marzipan cream cakes Puddings Milk pudding, custard, bread pudding, Christmas Fruit jelly, sorbet, cheesecake pudding, chocolate mousse, ice-cream Savoury snacks Nuts, Bombay mix, chevra, chana, ganthia, Popcorn, corn snacks poppadoms Sweets Chocolate, cocoa powder, halva, burfi with nuts, Mints, barley sugars, wine gums, Turkish delight, jelly Bounty, Snicker babies, chewing gum, lollipops Beverages Milk and milk drinks, Build-up, Complan, drinking Tea, coffee, fruit squash, lemon barley, Lucozade, soft chocolate drinks Miscellaneous Meat and yeast extracts such as Marmite and Bovril, Jam, honey, tomato ketchup, lemon juice, vinegar, marzipan, seeds such as sesame, tahini mustard, salad cream, herbs and spices

Sodium (1 mmol = 23 mg Na) and fluid levels may promote salt and fluid accumulation, while low intakes may lead to depletion and It is helpful to consider sodium and fluid dehydration. together since they are closely associated in the Dietary restriction should be implemented if body and the mechanisms for regulating their patients are hypertensive, oedematous or balance are integrated. The body’s sodium and require a fluid restriction for other reasons to an fluid status is a main determinant of blood pres- intake of <100 mmol sodium per day. sure and therefore is of clinical significance. The major sources of sodium in the UK diet People with renal failure lose their ability to are:3 adapt to changes in dietary sodium intake; high Pre-dialysis dietary management 243

Table 19.3 Low potassium diet sheet

Foods with a high potassium content Foods with a low potassium content

Cereals All-Bran, Bran Flakes, fruit and fibre, Raisin Splitz, Rice, noodles, pasta, bread, pitta bread, croissants, Sultana Bran puri, chapatti, Cornflakes, Rice Krispies, Flour, barley, sago, semolina, tapioca Meat substitutes Fresh soya bean products, vegetarian meat substitutes, bean milk Vegetables Ackee, artichokes, beetroot, mushrooms, spinach, Runner beans, cauliflower, carrots, cabbage, peas, celeriac, squash, tomatoes, parsnips, brussel sprouts, spring greens, asparagus, leeks, mangetout, courgette, okra, plantain and green bananas, dried pulses such sweetcorn as dried beans, red kidney beans, broad beans, butter beans, black-eyed beans, chick peas, lentils Fruit Avocado pear, banana, fresh blackcurrants or Apple, clementine, pear, satsuma, tangerine, passion redcurrants, dried fruit such as dried apricots, dried fruit, peach banana chips, currants, figs, prunes, raisins, sultanas Cakes and pastries Fruit cake, mince pies, Christmas cake, ginger nuts, oat Plain cake and biscuits, cream crackers cakes, rye crispbread, all biscuits and cakes containing dried fruit, nut and chocolate Puddings Bread pudding and Christmas pudding, desserts Fruit jelly, sorbet, cheesecake containing chocolate Savoury snacks Bombay mix, curu snacks, peanuts and raisins, potato Popcorn, corn snacks crisps, Twiglets, vegetable samosas, nuts Sweets Chocolate, all sweets containing chocolate or cocoa, Mints, barley sugars, wine gums, Turkish delight, jelly liquorice, toffees, fudge and other sweets containing babies, chewing gum, lollipops, fruit pastilles, sherbets, nuts, chocolate and dried fruit marshmallows Beverages Coffee, milk powder and drinks containing milk Tea (black or with a little milk), soft drinks such as tonic, powder such as Ovaltine, Horlicks, Complan, Build-up, lemonade, soda water, ginger beer, sparkling orange, drinking chocolate, milk shakes, cocoa powder, fruit fruit squash, lemon barley. Whisky, gin, vodka, brandy, juices unless exchanged for fruit, tomato juice, carrot rum juice, mango juice, strong ale (e.g. Guinness) Miscellaneous Tomato ketchup, tomato chutney, tomato puree, tomato Jam, honey, vinegar, Worcester sauce, mustard, sauce, meat and yeast extract, salt substitutes horseradish, sweet pickles, fresh herbs, spices containing potassium chloride (i.e. low-salt products)

• Bread and other cereals (35%) The remaining 22% is derived from a wide • Meat and meat products (26%) range of other foods. • Milk and milk products (8%) Concentrated sources of sodium are fairly • Pickles and sauces (7%). obvious because of their associated salty taste 244 Chapter 19 • Dietary management of kidney disease and include: ham, bacon, smoked fish, products dietary potassium should be restricted. For a canned in brine, cheese, salted butter, salted patient with some remaining residual renal foods (e.g. nuts, salted biscuits), yeast extract function, a more liberal potassium intake may spreads, stock cubes and bottled sauces. be allowed. Serum biochemistry and frequent Less obvious but significant sources of dietary assessments are necessary to prevent sodium include many meat and fish products, hyper- or hypokalaemia. canned and packaged soups, ‘instant’ foods and ready meals. The majority of manufactured Phosphorus foods now provide nutritional labelling in- formation which includes information on the Hyperphosphataemia is a problem encountered sodium content. 1.25 g salt (0.5 g sodium) or in the majority of patients. Low-phosphorus more per 100 g is a lot of salt; 0.25 g salt (0.1 g diets together with phosphate binders will be sodium) or less per 100 g is a little salt. needed to maintain pre-dialysis target range of <1.8 mmol/L.4

Dietary management of renal Sodium and fluid replacement therapy Soon after starting haemodialysis, urine output in the majority of patients will cease or reduce to very low volumes. Fluid and salt intakes Haemodialysis need to be restricted if hypertension and oedema are to be avoided. Patients often need Protein reminding that foods such as soup, milk pud- During haemodialysis there must be adequate dings, ice-cream have to be included within dietary protein to prevent malnutrition. An their fluid restriction. Many find that restricting intake of 1.0 g protein/kg IBW has been recom- their fluid allowance is the hardest part of their mended4 and appears adequate for stable diet. patients. However others advocate 1.2 g/kg day Useful tips for patients on a fluid restriction as a safer level, as this helps cover the needs of diet include: individuals with increased requirements. • Use a little salt in cooking and avoid adding Patients are provided with individual advice on salt to food at the table. the amount and type of protein they need to • Avoid or minimise the use of processed food. include within their diet. • Use herbs and spices to flavour food. • Measure the fluid allowance into a jug and Energy use throughout the day. • Use a small cup or glass instead of a mug. Energy requirements are not altered by • Drink half a cup each time. haemodialysis and an intake of 35 kcal/kg IBW • Rinse the mouth with water, gargle, but do is recommended.9 When assessing a patient’s not swallow. calorie intake a dialysis day and non-dialysis • Stimulate saliva production by sucking a slice day must be taken into consideration as there of lemon, sherbets or chewing gum. can be significant differences in terms of a patient’s dietary intake.

Peritoneal dialysis Potassium The aim of dietary restrictions is to maintain Continuous ambulatory peritoneal dialysis pre-dialysis blood levels within the range of (CAPD) is the most popular form of peritoneal 3.5–6.5 mmol/L.4 To avoid hyperkalaemia, dialysis, so most work on nutritional status and Dietary management of renal replacement therapy 245 requirements has been undertaken in patients retention. The restrictions depend on the on CAPD. amount of fluid dialysis is able to remove (Table 19.4). Protein Large amounts of protein are lost during the Transplantation dialysis process with an average daily loss of 15 g across the peritoneum.10 The amount of A well-functioning graft enables the restrictive protein lost can vary greatly between patients dietary regimens imposed in the pre-dialysis but during episodes of peritonitis protein losses and dialysis period to be relaxed. Nutrition increase significantly. To compensate for the though still remains an important aspect of high protein losses, a dietary protein intake of their care both in the peri-operative phase and >1.2 g/kg IBW is recommended. The conse- over the long term. quence of this is that a patient’s diet is often Both under- and overnutrition in the peri- higher in calories and phosphate. operative phase can adversely affect outcome in the kidney transplant recipient. Death rates are significantly increased in patients with a body Energy mass index (BMI) <18 kg/m2 and the risk of Glucose absorption from the dialysate across the graft failure rises with increasing BMI.11 Obese peritoneum will provide approximately 300 kcal recipients also suffer from significantly higher daily. The energy gain may be of benefit to some rates of delayed graft function, new-onset patients who are undernourished. A combined diabetes and higher mortality rates.12 Some (oral and dialysate) energy intake of >35 kcal/kg kidneys work immediately post transplant, but IBW is recommended. most take a few days to function. As the kidney function improves, a patient’s appetite and well- being will improve and previous restrictions Potassium can be relaxed once serum biochemistry has The aim is to maintain serum levels with the normalised. range of 3.5–5.5 mmol/L.4 Hyperkalaemia is less Longer term weight gain following trans- of a problem in CAPD than in intermittent plantation is well documemented. Average forms of dialysis and a more liberal potassium weight increases of 14% in the first year post intake is allowed. However, high levels are transplant have been reported.13 Dietary inter- monitored and a potassium restriction is initi- vention at an early stage has been shown to ated if required. limit weight gain successfully in this patient group.14 Hyperlipidaemia occurs in 25% of transplant Phosphorus patients, therefore a cardioprotective diet based Foods with high protein content commonly on healthy eating principles is appropriate.15,16 also have high phosphorus content. Consequently, the high protein diet recom- mended with CAPD will also contain more Acute renal failure phosphorus. To achieve the target range of <1.8 mmol/L4 a dietary restriction will usually The aims of dietary treatment for patients in be required and phosphate binders prescribed. acute renal failure (ARF) are the same as for all patients with kidney failure, that is to maintain nutritional status and limit the complications of Fluid and sodium renal failure. Malnutrition can develop rapidly Dietary intakes of both fluid and sodium will in this patient cohort and changes in fluid, need to be limited to prevent excessive fluid electrolyte and acid–base balance are more 246 Chapter 19 • Dietary management of kidney disease mono-unsaturated fatty acid; PDUO, previous urine day’s output; EAR, estimated required required energyReduced SFA Increased PUFA and MUFA Increase fibre energy use binder if Increased PUFA and MUFA (19–31 mmol/L) hyperkalaemia required Increased PUFA 2500 mg)and MUFA (31–45 mmol/L) volume as use binder if (31–45 mmol/L) use binder if medically indicated + UF capacity decreased SFA IBW) IBW) Protein (g/kg IBW) (kcal/kg Energy Fat CHO Phosphate (mg/day) Potassium (mmol/kg Sodium (mmol/day) Fluid Nutritional requirements for patients with chronic renal failure or on renal replacement therapy Table 19.4 Table Ideal high body biological weight; IBW, value HBV, saturated protein; fatty polyunsaturated SFA, acid; fatty PUFA, acid; MUFA, average requirements (national agreement). Based on ref. 9. CRF 0.6–1.0Haemodialysis 1.0–1.2 30–35 40% of 30–35 Peritoneal dialysis 1.2–1.5 Reduced SFA 30–35 50% of 30–35Transplant Reduced SFA 600–1000 1000–1400 30–35 EAR 1.0 Approx. 1.0 with 80–110 (1800– 1000–1400 If required, to a EAR 1.0 EAR – 80–110 80–110 500 EAR mL + PDUO 800 mL + PDUO Free Free 80–110 Free Vitamins and minerals 247 pronounced. Patients’ nutritional requirements haemoglobin and myoglobin of foods derived can be broadly categorised according to the dis- from animals, is relatively available and its order causing the kidney failure. ARF from a absorption is relatively unaffected by other food catabolic cause such as sepsis or trauma is items. The absorbability of non-haem iron pres- accompanied by increased protein turnover and ent in plant-derived foods depends on other dietary needs. Macronutrients requirements in dietary components such as tannins, phytates ARF caused by an obstruction or non-catabolic and fibre, making it less readily solubilised and event are not elevated. available than haem iron. Tea and eggs are Patients with catabolic ARF are often notable inhibitors of non-haem iron, whereas extremely ill. Mortality rates are high (40–80%) vitamin C can enhance the absorption. but appropriate feeding is associated with Good sources of haem iron are: improved survival.17,18 Each patient therefore • Red meat requires an individual dietary assessment and • Liver and offal meats. treatment plan based on the degree of catabol- ism, organs affected, biochemistry and renal Good sources of non-haem iron are: replacement therapy used. Artificial nutrition is frequently required, with daily assessment of • Bread and cereal foods made from UK forti- fluid status and biochemistry. fied white flour • Fortified breakfast cereals • Green leafy vegetables Vitamins and minerals • Pulses • Dried fruit • Nuts and seeds. Magnesium (1 mmol = 24 mg Mg)

Magnesium functions in many enzyme systems, Vitamins such as those involved in decarboxylation or phosphate group transfer and energy release. It Vitamins regulate the metabolic pathways of plays a vital role in skeletal development, pro- protein, carbohydrate and fat, and end stage tein synthesis, muscle contraction and neuro- renal failure alters the serum levels, body stores transmission. Metabolically, it is closely linked and function of many of these vitamins. with calcium. Homeostatasis is largely con- Deficiencies also occur due to the dietary restric- trolled by the kidneys. tions imposed on patients. The B vitamins – B6, folic acid and B12 – are more likely to be affected by drug interactions. Iron (1 mmol = 56 mg Fe) These vitamins may improve poor erythro- The major role of iron is as an oxygen carrier poietic responses in patients on dialysis. in haemoglobin in the blood and myoglobin in An average of 125 mg vitamin C is lost dur- muscle. It is also required for many metabolic ing each dialysis session and supplementation is processes, including the citric acid cycle and often required, especially in malnourished amino acid metabolism. patients. However a supplement of 200 mg The human body contains 3–5 g iron, about vitamin C may increase oxalate levels. Oxalate two-thirds of which is in haemoglobin. deposits as crystals in soft tissues such as muscle The UK recommended nutrient intake (RNIs) and vital organs and may increase the risk of for iron for adults are are follows: myocardial infarction, muscle weakness and bone disease. • Men >19 years: 8.7 mg/day Vitamins A, D, E and K are the fat- • Women 11–50: 14.8 mg/day soluble vitamins and vitamin supplements con- • Women >50: 8.7 mg/day. taining these are contraindicated in patients Dietary iron exists in two forms: haem and non- with renal failure. Vitamin A metabolites are haem. Haem iron, which is contained in the excreted poorly and can accumulate over time. 248 Chapter 19 • Dietary management of kidney disease

High-dose vitamin E supplementation may be a There is no single or standard way of assessing potential risk factor affecting the clotting mech- nutritional status. Nutritional status is a anism. Vitamin K is contraindicated unless a dynamic entity reflecting physiological require- patient is on chronic antibiotic treatment.19 ments, nutritional intake, body composition and function and all these have to be consid- ered and the findings interpreted in conjunc- Malnutrition tion with one another.

There have been many papers published since Dietary considerations the early 1980s on the consequence of malnu- These focus on the extent to which dietary trition, including: intake is likely to meet nutritional needs. Factors to be assessed include: • Increased morbidity • Delayed wound healing • Current food and fluid intake • Increase risk of infection • Duration and severity of any changes in • Electrolyte imbalance appetite and oral intake • Prolonged hospitalisation • Presence of factors that may be affecting food • Increased mortality. and fluid intake. In the renal population various factors lead Methods of dietary assessment include asking to a risk of protein–energy malnutrition, includ- a patient to recall in as much detail as possible ing existing malnutrition, uraemia, depression, everything consumed in the last 24 hours or co-existing gastrointestinal disease such as asking patients to keep food diaries, usually for gastroparesis, cancer, heart failure, anorexia, 5–7 days, which are then assessed. financial constraints, increases in nutritional losses due to proteinuria, and losses during Anthropometric consideration dialysis. Routine nutritional assessment and individual advice is essential to identify patients Anthropometric methods are used to assess at risk of developing malnutrition.20 body composition in living people. Anthro- pometric parameters reflect both health and nutritional status and can predict performance, Assessment of nutritional status health and survival. For practical purposes, body composition can be considered to be comprised Assessment of nutritional status means deter- of lean body mass, fat stores and body water. mining the extent to which an individual’s Ways in which these can be measured are sum- nutritional needs have been or are being met. marised in Table 19.5.

Table 19.5 Anthropometric measurements of body composition

Protein status Fat stores Body water

Mid-arm muscle circumference Triceps skin fold thickness Bioelectrical impedance Grip strength Body mass index Biochemistry Nitrogen balance Fluid balance charts Plasma proteins Rapid weight changes Plasma urea Girth (asites) Pitting oedema Malnutrition 249

Body mass index support in the form of enteral or parenteral nutrition. Height and weight measurements are easy to perform and BMI can be calculated using the following formula: Nutritional supplementation

BMI = weight (kg)/height (m)2 Many sip-feed supplements are available on an ACBS (Advisory Committee on Borderline The BMI reflects body fat stores and has import- Substances) prescription for severe under- ant predictive values in terms of morbidity nutrition as detailed in the British National and mortality in those classified as underweight Formulary (BNF). Some are designed to meet or obese:21 higher energy or protein needs or with particu- lar clinical indications such as renal disease. • Severely underweight <16 One potential problem with nutritional supple- • Underweight <18.9 ments is that few of the products are suitable for • Normal range 19–24.9 vegans (or people who strictly follow Vegetarian • Overweight 25–29.9 Society guidelines) or for Kosher diets. Oral sup- • Obese class I 30–34.9 plements should be tried initially, although • Obese class II 35–39.9 their efficacy has been shown to be greatest in • Obese class III 40–60. those with a BMI <20 (Table 19.6). 23

Subjective global assessment Enteral feeds Subjective global assessment is an assessment of nutritional status. It is based on the patient’s Feeding enterally is superior to the parenteral history and physical examination. The medical route in terms of physiology, immunology and history involves asking the patient questions cost, and has numerous clinical advantages such about: weight changes, dietary intake, gastro- as helping to maintain normal intestinal func- intestinal symptoms and functional impair- tion and structure.24 Mechanical obstruction, ment. The physical evidence includes signs of: prolonged ileus and cardiovascular instability are loss of subcutaneous fat, muscle wasting, the only absolute contraindications to enteral oedema and ascites. These are observed at nutrition.24 Guidelines on the management of several locations on the body, including the enteral nutrition in the adult patient have been face, clavicle, back, hands, arms, legs and produced by the British Society of Gastro- ankles. The overall rating depends on the scores enterology.25 Specialist renal feeds with reduced given, from normal to mild malnutrition to electrolyte content are available. These are useful severely malnourished.22 where the control of serum phosphate, potas- sium or fluid balance proves difficult. Complications of enteral tube feeding Nutritional support include:26 Nutritional support does not just mean the use • Aspiration – This may occur with no obvious of supplements or enteral/parenteral nutrition. vomiting or coughing, and pneumonia can The first step in the process of providing nutri- develop silently. tional support is simple dietary advice focused • Gastrointestinal symptoms. Nausea occurs in on improving the quality and quantity of food 10–20% of patients and abdominal bloating which may be sufficient to correct or avert the and cramps from delayed gastric emptying problem. Food enrichment (with fat, sugar are also common. Diarrhoea occurs in up to and/or protein) may help improve nutrient 30% of enterally fed patients; this can create density. Some people may need further support serious problems from nutrient, fluid and in the form of sip-feed and other supplements. electrolyte losses, and from infected pressure Only a few will require artificial nutritional sores and general patient distress. 250 Chapter 19 • Dietary management of kidney disease Nutritional content of oral dietary supplements Table 19.6 Table Nutritional supplementQuantity Ensure Energy Plus(kcal) FortisipProtein (g) Fresubin 330Fat (g) 220 mL Enlive PlusCHO (g) 200 Provide mL 300 13.8Sodium (mmol) 200 mL ComplanPotassium (mmol) Energy 12.0 11.5 300 Renilon 7.5 220 10.8Phosphate (mg) mL 11.3 44.4 Maxijule 105CHO = carbohydrate. 202 mg 11.3 210 11.6 330 200 mg mL 36.8 Calogen 7.0 Formance 10.6 108 11.6 7.0 230 mL 37.6 Extra Nepro 250 125 100 1.05 mL 0 71.9 0.92 7.5 244 100 g 24.2 2.4 55 2.4 9.4 0 250 100 mL 80 5.7 11.8 113 31.3 g 9.4 400 9.0 270 3.2 100 0.4 mL 25.0 0 12.5 466 7.5 0 0 0 167 0 0 0 <0.4 200 0 4 50 3.96 4.67 0 27.1 3.91 7.0 4.97 3.67 22.2 2.72 99.4 9.6 69.0 Malnutrition 251

• Blocked feeding tubes. Tubes can block easily, • Physiological – rise in serum bilirubin, sec- especially if they are not flushed with fresh ondary to reduced bilirubin binding capacity tap, cooled boiled or sterile water before and and increased free circulating bilirubin due to after every feed or medication. Any drugs free fatty acids from the lipid emulsion of the administered through a tube should ideally parenteral nutrition. Excessive glucose can be suspensions rather than syrups. Hyper- lead to a fatty liver. osmolar drugs, crushed tablets, potassium • Infections – either secondary to the catheter and iron supplements are particularly likely or non-catheter concurrent infections. to cause problems. A tube can often be unblocked by flushing with warm water. Intradialytic parenteral nutrition Intradialytic parenteral nutrition (IDPN) is given Parenteral nutrition whilst a patient is undergoing haemodialysis Parenteral nutrition is a method of providing using the same vascular access. This may nutritional support to an individual whose improve nutritional status and protein kinet- gastrointestinal tract is not functioning or is ics.27 Parenteral formulas containing approx- inaccessible. Nutrients are delivered directly into imately 65–70 g protein and 1000 kcal (mainly the circulatory system via a dedicated venous glucose) can be safely delivered during catheter. Although parenteral nutrition is essen- haemodialysis treatment. Glucose monitoring tial and a potentially life-saving therapy, it is during treatment is essential to prevent hyper- expensive and carries life-threatening complica- glycaemia. The fluid balance can be adjusted tions (sepsis and metabolic disorders), so must accordingly. IDPN still remains a controversial be monitored and administered correctly. The mode of nutritional support, but an evidence- main complications of parenteral nutrition are: based evaluation by Foulks28 showed it to be of benefit. The cost of IDPN is 10 times that of • Metabolic – fluid overload, hyperglycaemia enteral nutrition or oral supplementation. and electrolyte abnormalities.

C CASE STUDY

Mr Smith is a 63-year-old man referred to the dietetic outpatient service with stage 4 chronic kidney disease for a renal diet. His biochemistry is within expectable ranges, with no reported weight loss but he is hypertensive.

Q1. What advice would you give him?

At his next review his serum phosphate has increased (PO4 2.10 mmol/L) and the consultant has started him on phosphate binders.

Q2. What advice would you give him now? Twelve months later his eGFR has dropped to 14. His serum potassium has increased (K 6.4 mmol/L).

Q3. What further dietary measures would be needed? As Mr Smith approaches dialysis his weight begins to decrease and the doctor has requested some nutritional supplements for a renal patient.

Q4. Which supplements would you suggest? 252 Chapter 19 • Dietary management of kidney disease

References 15. Lawrence IR, Thompson A, Hartley GH et al. The effect of dietary intervention on the management of hyperlipidemia in British renal transplant 1. Berlyne GM. A Course in Renal Disease, 2nd edn. patients. J Ren Nutr 1995; 5: 73–77. Oxford: Blackwell Scientific Publications, 1968. 16. Nelson J, Beauregard M, Gelinas M et al. Rapid 2. Klahr S, Levey AS, Beck GJ et al. The effects of improvement of hyperlipidemia in kidney trans- dietary protein restriction and blood-pressure plant patients with a multifactorial hypolipi- control on the progression of chronic renal dis- demic diet. Transplant Proc 1988; 20: 1264–1270. ease. N Engl J Med 1994; 330: 878–884. 17. Rainford DJ. Nutritional management of acute 3. National Diet and Nutrition Survey (NDNS). renal failure. Acta Chirurg Scand 1981; 507 Adults aged 19–64 years. Medical Research (Suppl): 327–329. Office/Office for National Statistics, 2003/2004. 18. Bartlett RH, Mault JR, Dechert RE et al. 4. Renal Association. Treatment of Adult Patients with Continuous arteriovenous hemofiltration: im- Renal Failure. Recommended Standards and Audit proved survival in surgical acute renal failure? Measures, 3rd edn. London: The Royal College of Surgery 1986; 100: 400–408. Physicians, 2002. 19. Rocco MV, Makoff R. Appropriate vitamin ther- 5. Kopple JD, Monteon FJ, Shaib JK. Effect of energy apy for dialysis patients. Semin Dial 1997; 10: intake on nitrogen metabolism in nondialyzed 272–277. patients with chronic renal failure. Kidney Int 20. Malnutrition Advisory Group of the British 1986; 29: 734–742. Association for Parenteral and Enteral Nutrition 6. Slatapolsky E, Delmez JA. Pathogenesis of sec- (BAPEN). Guidelines for the Detection and ondary hyperparathyroidism. Nephrol Dial Management of Malnutrition. Maidenhead: BAPEN, Transplant 1996; 3 (Suppl 11): 130–135. 2000. 7. Rufino M, Bonis ED, Martin M et al. Is it possible 21. de Onis M, Habicht JP. Anthropometric reference to control hyperphosphataemia with diet, with- data for international use: recommendations out inducing protein malnutrition? Nephrol Dial from a World Health Organization Expert Transplant 1998; 13 (Suppl 3): 65–67. Committee. Am J Clin Nutr 1996; 64: 650–658. 8. Bansal VK. Potassium metabolism in renal failure: 22. Detsky AS, McLaughlin JR, Baker JP et al. What is nondietary rationale for hyperkalaemia. J Ren subjective global assessment of nutritional status? Nutr 1992; 2: 8–12. J Parenter Enteral Nutr 1987; 11: 8–13. 9. European Dialysis and Transplant Nurses 23. Stratton RJ, Elia M. A critical systemic analysis of Association/European Renal Care Association the use of oral nutritional supplements in the (EDTNA/ERCA). Nutritional Guidelines 2001. community. Clin Nutr 1998; 18 (Suppl 2): 29–84. www.edtna-erca.org (accessed 7 November 2001). 24. McClave SA, Snider HL, Spain DA. Preoperative 10. Blumenkrantz MJ, Gahl GM, Kopple JD et al. issues in clinical nutrition. Chest 1999; 115: Protein losses during peritoneal dialysis. Kidney 64S–70S. Int 1981; 19: 593–602. 25. Kirby DF, Teran JC. Enteral feeding in critical 11. Chertow GM, Lazarus JM, Milford EL. Quetelet’s care, gastrointestinal diseases and cancer. index predicts outcome in cadaveric kidney Gastrointest Endosc Clin N Am 1998; 8: 623–643. transplantation. J Ren Nutr 1996; 6: 134–140. 26. Stroud M, Duncan H, Nightingale J. Guidelines 12. Holley JL, Shapiro R, Lopatin WB et al. Obesity as for enteral feeding in adult hospital patients. Gut a risk factor following cadaveric renal transplant. 2003; 52 (Suppl 7): vii1–vii12. Transplantation 1990; 49: 387–389. 27. McCann L, Feldman C, Hornberger J et al. Effects 13. Przygrodzka F, Rayner HC, Morgan AG, Burden of intradialytic parenteral nutrition on delivered RP. Change in nutritional status after successful Kt/V. Am J Kidney Dis 1999; 33: 1131–1135. renal transplantation. J Ren Nutr 1992; 2: 18–20. 28. Foulks CJ. An evidence-based evaluation of intra- 14. Patel MG. The effect of dietary intervention on dialytic parenteral nutrition. Am J Kidney Dis weight gains after renal transplantation. J Ren 1999; 33: 186–192. Nutr 1998; 8: 137–141. 20

Travelling and vaccines

Zoe Thain

This chapter discusses issues in relation to travel sometimes prove difficult, however, to accom- for the patient with renal disease based in the modate a specific destination with specific UK. It begins with general considerations to be travel dates, and patients may have to be quite taken when booking holidays for different flexible. Patients awaiting transplant who go groups of renal patients, and goes on to exam- abroad may be suspended from the transplant ine both general travel health advice and spe- list for the duration of their holiday, depending cific advice relating to travelling abroad with on the destination. medication. It also addresses the requirements for malaria prophylaxis and travel vaccinations. Patients have various reasons for travel. They Haemodialysis may wish to visit relatives abroad, may be required to travel for business reasons, or may Patients can usually choose their travel destina- just want to go on holiday. The key message is tion, although this will have to be close to a that with appropriate planning and advice, kid- dialysis unit that can accommodate visitors. It is ney patients can travel and that destinations often easier to find dialysis space abroad than in worldwide are possible. the UK, as NHS hospital units tend to work at full capacity. Private units in the UK may pro- vide a suitable option, usually at a similar cost to NHS units. All holiday dialysis sessions in the Planning holidays UK are paid for by the patient’s purchasing authority. The first priority when a renal patient wants to There are various holiday directories on the go on holiday is to assess if they are fit to travel. Internet to help patients and co-ordinators All patients, even transplant patients, will locate suitable dialysis units. Eurodial is a help- require a letter from their renal unit confirming ful guide to dialysis units for patients travelling this in order to obtain suitable travel insurance. to Europe, and Holiday Dialysis International This also provides a first point of contact for the and Global Dialysis will locate units worldwide, patient for advice on reducing illness and health including cruises. Recommendations for suit- risks whilst travelling, including advice on suit- able units may also be provided by other able malaria prophylaxis and vaccinations if patients, based on their own travel experiences. required. There may be a charge per dialysis session Most renal units now have a designated depending on the destination. Most countries member of staff with the role of holiday co- that accept the European Health Insurance Card ordinator who can assist with arranging the (EHIC) within the European Union have a recip- holiday, and suggest suitable destinations. rocal arrangement with the UK; however a por- Whilst last-minute bookings are certainly not an tion of the cost may still need to be paid, and option for a holiday-dialysis patient, most des- some private clinics may not accept the EHIC. tinations are possible with good planning. It can For countries not covered by the EHIC, charges

253 254 Chapter 20 • Travelling and vaccines may vary from between £150 and £200 for each require a letter from their renal unit to explain dialysis session in Turkey, to £500 per session in the need for excessive hand luggage and that it the USA. Patients themselves have to pay for is required for medical treatment. these dialysis costs. The need to carry out an exchange during Patients should be prepared for differences in the journey will depend on the itinerary, the equipment and procedures used in units in although it is always advisable to carry one dial- other countries, and in some cases may have to ysis bag in hand luggage in case of delays. The take their own dialysers and medications for peritoneal dialysis nurse can advise on a suitable dialysis, such as heparin and erythropoietin. exchange plan whilst travelling, and give advice Transport between hotel and dialysis unit will on where to carry out exchanges to reduce the not necessarily be provided, and patients will risk of peritonitis. Renal units may provide need to pay for and organise this themselves. It patients with antibiotics to take with them in is essential, therefore, that patients are provided case of peritonitis, particularly if they are trav- with full details of when their dialysis sessions elling to places that are not in close proximity will be before travel. to medical assistance, or are on a cruise ship. Recent blood results, including MRSA, HIV, Appropriate equipment for administration and hepatitis B and hepatitis C status will be full instructions for treatment should also be required by the holiday-dialysis unit. These are provided. usually requested to be taken within four weeks of travel. When planning trips to high-risk areas for blood-borne viruses, it must first be estab- Transplant patients lished that the base unit will be able to accom- modate the traveller to dialyse in isolation on Transplant patients are advised not to travel to return to the UK. Each renal unit in the UK will countries where the risk of catching an infec- have a policy for the isolation of patients tion is high; otherwise they are not restricted in returning from overseas dialysis. their choice of destination. Precautions should always be taken to reduce the risk of infection whilst on holiday, such as drinking only bottled Peritoneal dialysis or boiled water, and avoiding salads and ice- cubes. Transplant patients are at increased risk Patients who require peritoneal dialysis have a of skin cancer because of immunosuppressant greater freedom to travel, the only restrictions drugs,1 so sun exposure should be kept to a min- being whether the dialysis fluids can be deliv- imum. It is vital that sun protection measures, ered to the destination of choice, and that they including the use of high-factor sun block, are can be stored appropriately in the holiday taken. accommodation selected. Delivery is arranged by the manufacturer of the particular fluid, and renal units will help to co-ordinate this for General travel health advice patients. It may take up to three months to make the necessary arrangements; the notice required for delivery of fluids is generally 2–4 Any traveller should consider taking a basic first weeks for the UK, 8 weeks for Europe and 12 aid kit containing items such as plasters and weeks for worldwide destinations. Patients dressings, antiseptic, sun block, insect repellent, should call the destination a few days before and water-sterilisation tablets. Other medicines travel to ensure supplies have arrived safely, and that can be bought over the counter (OTC), know who to contact if there are any problems which may only be required occasionally, with supplies during the holiday. Patients may should also be considered by the traveller with be responsible for transporting their machines kidney disease. These should be purchased and ancillary products themselves, and will before travel so that suitable OTC medicines can General travel health advice 255 be recommended. It should be confirmed that needles and other equipment may be in short there are no interactions with prescribed medi- supply or not always sterilised, patients may cines, that doses and medicines are appropriate consider taking an emergency medical travel kit. for the degree of renal impairment, and that These can be purchased from pharmacies and they are not likely to worsen renal function. The contain sterile needles, syringes, and dressing exact content of the kit will be determined by packs, which can be handed to a doctor or nurse the type of traveller, the destination and likely for use in an emergency. activities; factors which may affect whether a person will develop a travel-associated illness. Over-the-counter medicines suitable for renal Traveller’s diarrhoea patients are listed in Table 20.1. 2 Apart from their usual medications it may be Traveller’s diarrhoea is the most common illness necessary to provide renal patients with medi- contracted abroad by travellers generally, with cines for self-treatment in emergency situations, incidence estimated to be between 30 and 50%. particularly if travel is to remote areas or far It is usually defined as the passage of three or from medical facilities. For example, renal units more unformed stools in a 24-hour period, with may supply continuous ambulatory peritoneal at least one other symptom of enteric disease dialysis (CAPD) patients with antibiotics, based such as nausea, vomiting, abdominal pain, on their own protocol, for the initial treatment fever, faecal urgency, or blood or mucus in the of peritonitis. These may be required if patients stools.3 Traveller’s diarrhoea, along with hepat- are travelling outside Europe, where appropriate itis A and typhoid, is contracted through antibiotics may not be readily available, or on contaminated food and water. The risk of con- cruise ships which do not carry supplies of such tracting diarrhoea can be reduced by taking the drugs on board. Full instructions should be following simple precautions:4 given to the patient, so that they understand when and how to use these medications. • Wash your hands after using the lavatory and For travel to resource-poor areas where before handling and eating food.

Table 20.1 Over-the-counter medicines for renal patients

Ailment Recommended Avoid Other information

Headache Paracetamol Aspirin Effervescent tablets contain sodium so Co-codamol Ibuprofen avoid Coughs and colds Paracetamol Decongestants Steam inhalation, menthol or olbas Simple linctus oil may help congestion Muscle aches Ralgex Topical and oral NSAIDs Deep heat Indigestion Gaviscon Aluminium- or Gaviscon for short-term use only as magnesium-containing contains sodium indigestion remedies Hayfever Antihistamines Nasal sprays and eye drops may also be effective Constipation Senna Fybogel (if fluid restricted) Seek medical advice if symptoms persist Diarrhoea Loperamide Oral rehydration salts Seek medical advice if symptoms are severe or persist 256 Chapter 20 • Travelling and vaccines

• Use bottled water or boiled or sterilised tap Insect bites water for drinking, washing food or cleaning teeth in countries where sanitation may be The risk of infection spread by insects, includ- poor. ing malaria, can be reduced by good protection • Avoid ice in drinks and ice used to keep food against insect bites. cool. • Hot tea, coffee, wine, beer, carbonated water Clothing and soft drinks, and packaged juices are usu- ally safe. Travellers should be advised to wear long- • Eat freshly cooked food which is piping hot. sleeved clothing, long trousers, and socks out of • Avoid food which has been kept warm. doors between dusk and dawn. • Avoid salads or uncooked fruit or vegetables, unless you can peel or shell them yourself. Insect repellent • Avoid unpasteurised milk, ice-cream and dairy products. Of the products available on the market DEET • Avoid food from unreliable sources such as (diethyltoluamide)-based products are the most street vendors. effective, and should be used first line for travel • Avoid raw seafood and shellfish. to malarious areas. They are available in differ- • Avoid food likely to have been exposed to ent concentrations for use on skin or clothing. flies. There have been safety concerns as DEET can be absorbed after application to the skin, but as Most cases of traveller’s diarrhoea are mild, long as recommended concentrations of lasting a few days, and most travellers will 20–50% are not exceeded and it is used as recover with symptomatic treatment. Oral rehy- directed by the manufacturer, then clinical dration solutions contain both sodium and experience shows it is safe to use.6 It should potassium, so should be avoided in people with only be used on exposed areas of the skin and kidney disease, although travellers should be applied with care to the face, avoiding the advised to maintain their usual fluid intake. The mouth and eyes. It should not be used on bro- antimotility agent loperamide is suitable for use ken or irritated skin. Hands should be washed by renal patients and will reduce symptoms of after application and the repellent should be faecal frequency and stomach cramps. However, removed with soap and water when no longer constipation can develop if loperamide is taken required. Use of sunscreen which contains repel- at maximum dose for more than a day or so. lent should be avoided as repeated administra- There is also a risk that infection may be pro- tion may result in excessive use of DEET. longed by the use of antimotility agents due to retention of the organism, so loperamide should be avoided if there is fever or bloody Insecticides diarrhoea. Quinolone antibiotics such as Clothes and mosquito nets can be treated with ciprofloxacin have been used for the treatment an insecticide such as permethrin. Sprays, plug- of traveller’s diarrhoea.3 They may be carried by in devices and coils can also be used to rid those considered to be most at risk, such as sleeping areas of insects. Mosquito nets should be immunosuppressed patients, for immediate self- used unless the room has screened windows and treatment, although this is not a licensed indi- doors. Air-conditioned rooms are also advised. cation. Doses of quinolone antibiotics need to be adjusted according to renal function.5 Medical advice should be sought if symptoms Treatment of bites do not improve within a few days, if there is Bites and stings are commonly experienced by blood in the stools, or if a fever or confusion travellers, although the response may vary from develops. a mild allergic reaction to anaphylaxis. Most Travelling abroad with medication 257 bites can be treated with application of a mild whereas promethazine, which induces a high topical steroid cream to reduce swelling and oral degree of drowsiness, may be useful for night- antihistamines to relieve itching. Bites should time journeys. be kept clean and not scratched, to reduce the risk of infection. Oral cortico steroids may be a consideration if severe reactions are likely, and Deep vein thrombosis epinephrine (adrenaline) should be carried by those who have previously experienced ana- The risk of deep vein thrombosis (DVT) in rela- phylaxis with bites.3 tion to travel can be reduced by regular flexing of the ankles to encourage blood flow in the lower legs, avoiding alcohol and maintaining Sun protection adequate fluid intake. Additional measures may be considered for Overexposure to the sun can cause sunburn and those at high risk of DVT such as those with a heatstroke. General measures such as covering history of DVT, recent surgery or heart disease, up with suitable clothing and wearing a wide- pregnancy or hormonal medication, malig- brimmed hat and sunglasses that block UV radi- nancy, haematological disorders, varicose veins, ation should be taken. Staying in the shade and obesity, dehydration, or age over 40 years.3 avoiding direct sun between 11 am and 3 pm Well-fitting compression stockings may will also help reduce exposure. Patients should reduce the risk of DVT in high-risk patients, as be advised to take extra care if they are also may the use of low-molecular-weight heparin. A prescribed medications (e.g. doxycycline) that suitable regimen should be discussed with med- sensitise the skin to UV light. The correct use of ical staff as dose adjustment is required for renal appropriate suncream is vital, and travellers impairment. British Medical Association guide- should use suncream with a sun protection fac- lines7 support the view that aspirin should not tor (SPF) of at least 15.3 The National Kidney be used for the prevention of DVT in travellers Federation advises transplant patients to use SPF due to insufficient evidence. 25 or higher. The appropriate amount to apply is often underestimated; 100 mL will cover the body approximately three times. Suncream Travelling abroad with medication should be applied at least 30 minutes before sun exposure and reapplied frequently, at least every 2 hours. Renal patients are often required to take a Calamine lotion or cream can be used to multitude of different medications. When plan- soothe mild sunburn, and paracetamol taken if ning a holiday, particularly abroad, it is essen- analgesia is required. tial that preparations are made well in advance of travel in order to arrange sufficient supplies. Patients should see their GP to arrange a pre- Travel sickness scription to cover the holiday period, plus a few extra days in case of delays. British GPs can Medications used to prevent motion sickness prescribe a maximum of three months’ supply include cinnarizine, hyoscine and prometh- on an NHS prescription for overseas travel. After azine. All are suitable for use by renal patients this time the individual ceases to be registered at usual doses.5 They differ in duration of action with the GP.8 and side-effects which may help determine the It is advisable that patients obtain a letter most appropriate agent to use. For long journeys from their GP detailing all medications (with cinnarizine may be useful as it has a long dura- generic names not just brand names), any tion of action, whereas hyoscine has a short equipment required (such as needles and duration. Cinnarizine causes least drowsiness, syringes), and the medical conditions for which 258 Chapter 20 • Travelling and vaccines they are being used, to confirm the patient’s down into smaller containers, but this could need for the medication. Although not compul- lead to problems at customs. It will also prove sory, it is likely to prove helpful when going useful if the patient needs to consult a doctor through customs. As a minimum patients overseas, that full instructions and information should at least take a copy of their prescription. leaflets for their medications are available. For Many countries, particularly those outside extra protection, travellers could consider stor- Europe, such as India, Pakistan, Turkey and ing items in a sealable plastic box or, if this is some Middle Eastern countries, have lists of too bulky, then in resealable plastic bags to medicines which they do not allow to be protect against moisture. Items such as erythro- brought in. These might even include medicines poietin or insulin which need to be kept cool available over the counter in the UK as the legal can be carried in small cool bags or boxes, status of medicines varies between countries. although care should be taken to avoid direct The relevant Embassy in the UK should be con- contact with ice packs. For long flights, airlines tacted well before travel, to ensure the patient may provide cool storage. Documentation, such will have no problems taking the medicines as a letter from the GP, will be required for they need. The UK Foreign and Commonwealth patients to carry syringes and needles in their (FCO) website has contact details for Embassies hand luggage (Box 20.1). and High Commissions in the UK. Consideration should also be given to how the medication will be stored at the holiday des- tination, particularly for long trips. For items Controlled drugs

For any medication classed as a controlled drug in the UK, a Home Office licence may be Box 20.1 Travel documentation which may be required to take it abroad. The HM Revenue and required by a renal patient Customs leaflet, ‘Taking medicines with you when you go abroad’, available via www.hmrc. gov.uk, gives a list of permitted allowances for • Doctor’s letter – detailing medications, medical controlled drugs. If the patient needs to take equipment and medical conditions more than the permitted allowance, they must • Copy of prescription apply in writing to the Home Office for a licence • Home Office licence – if required for con- giving the following details: trolled drugs • European Health Insurance Card (EHIC) – • Patient’s name, address, and date of birth replaced the E111 as from 1 January 2006. • Country/countries of destination This entitles the traveller from the UK to • Dates of departure from and return to UK reduced-cost, sometimes free, medical treat- • A letter from the prescribing doctor confirm- ment that becomes necessary, because of ill- ing the drug details (name, form, strength ness or accident. It also covers treatment and total quantity). which may be needed for a chronic disease This should be done at least two weeks before or pre-existing illness. Arrangements will need travel to allow time for the licence to be issued. to be made in advance for dialysis • Travel insurance – must cover for pre-existing medical conditions (the National Kidney Packing and storage Federation can recommend suitable insurance) and/or have specific health insurance All medication should be stored in hand lug- • Letter for airline if carrying excess hand lug- gage, so that it is readily accessible, with each gage (CAPD patients) item in a correctly labelled container, as issued • Copy of blood results and medical information by the pharmacist. It may be tempting for the – if requested by the holiday dialysis unit patient to carry loose tablets or pack medication Malaria 259 that require refrigeration, patients should con- guidelines formulated by the Health Protection sider booking a room with a fridge, using a cool Agency (HPA) Advisory Committee on Malaria box, or if necessary, storing such items in the Prevention (ACMP) for UK Travellers. 9 A full coolest, darkest area possible. The stability of update of the current guidelines was carried out medicines exposed to extremes of temperature in 2006, but specific updates and notices are has not been studied, so patients should con- available on the ACMP web pages (http://www. sider discarding unused medicines after return- hpa.org.uk/publications/2006/Malaria/Malaria_ ing from a long trip to particularly warm or cold guidelines.pdf). There are separate guidelines climates.8 produced by the ACMP for advising long-term travellers (longer than six months) on malaria prophylaxis.10 Purchase of medicines overseas Taking appropriate chemoprophylaxis is only one aspect of malarial prevention. The guide- Patients should be advised to take all their med- lines give four essential ABCD steps,9 which are ications with them, including those prescribed the same throughout the world, and relevant to and any OTC medicines which are likely to be any traveller regardless of their medical history. required. This is particularly important if travel is to remote areas or developing countries where counterfeit or poor-quality medicines may be Awareness of risk supplied.8 Specific medicines to treat chronic conditions may not be marketed in other coun- The risk of malaria is determined by the destina- tries or availability could be unreliable. There is tion, duration of the visit, and likely degree of also the problem of communication, as trav- exposure due to intended activities and style ellers may be unable to adequately express their of travel. The risk in a particular country may requirements, or may not be able to follow also vary depending on the time of year, and information about how to take medicines they between rural and urban destinations. have been supplied or prescribed whilst abroad. Particularly at risk are ethnic groups in the UK If purchase of medicines is unavoidable, then (or long-term visitors to the UK) who visit reliable retailers recommended by an Embassy friends and relatives in their country of origin. or Consulate should be used. These travellers may assume they still possess some immunity, which actually fades quickly, and so do not take adequate, if any, preventat- ive measures. Malaria

Malaria is a significant health risk of travel Bites by mosquitoes: prevent or avoid abroad with approximately 2000 cases reported in the UK each year. Almost all malaria deaths, Taking effective measures to reduce mosquito on average nine per year in the UK, are pre- bites will significantly reduce the risk of con- ventable.9 Most cases of malaria occur in those tracting malaria, and will also protect against who fail to comply with malaria prophylaxis, so other insect-borne infections. Bite prevention it is essential that renal patients are counselled measures should be taken by all travellers as no as to the importance of taking another drug in malaria prophylaxis regimen will provide com- addition to their usual medications. The degree plete protection. of renal failure and the drugs taken by renal patients should be taken into account when considering the most suitable prophylaxis for a Compliance with appropriate chemoprophylaxis particular patient. Healthcare workers who advise travellers Patients should be counselled on the import- about malaria prophylaxis should refer to the ance of taking the medication regularly and for 260 Chapter 20 • Travelling and vaccines the correct duration. Malaria is as likely to occur attention immediately and mention their poss- in those who take prophylaxis irregularly as in ible exposure to malaria if they develop any of those who take no prophylaxis at all. Weekly the following symptoms: regimens should be started at least one week • Fever (three weeks for mefloquine) before travel. • Flu-like illness Malarone (atovaquone/proguanil) can be started • Backache one to two days before travel into an endemic • Diarrhoea area. This will allow identification of any • Joint pains adverse effects before travel, and provide suffi- • Sore throat cient time to switch to an alternative medica- • Headache. tion. Travellers should be advised of possible side-effects and to seek advice if severe. Antimalarials should be taken while in a mal- Choice of chemoprophylaxis arious area and for four weeks after leaving (one week for Malarone). This will protect against There are currently six prophylactic regimens malaria contracted at the end of travel. described in the ACMP guidelines for UK trav- Daily doses should be taken at the same time ellers.9 These are given, along with usual adult each day, and weekly doses at the same doses, in Table 20.2. Doses of prophylactic anti- time each week. Taking after meals will help malarials for children, based on weight, can also reduce side-effects and provide a prompt to be found in the ACMP guidelines, and in the aid compliance. Travellers should obtain their BNF for children. full course of medication before travel, as the The ACMP guidelines give the recommended strength of antimalarials abroad may differ prophylaxis regimen for specific areas and coun- and could therefore provide inadequate tries based on geographical resistance patterns. protection. Also, where possible, there are alternative regi- mens suggested for those unwilling or unable to follow a recommended regimen. In some Diagnose malaria swiftly and obtain instances there are several regimens of equiva- treatment promptly lent efficacy. The choice between them then is determined by the particular circumstances of Early symptoms of malaria can be non-specific. the traveller. The guidelines consider the Although symptoms usually occur within three importance of balancing the risk of malaria and months of being bitten by an infected mos- the risk of adverse reaction to antimalarials, quito, malaria can present at any time between including observing the contraindications to one week and one year after exposure.11 the use of specific antimalarials. They also Travellers should be advised to seek medical include recommendations for prophylaxis in

Table 20.2 Prophylactic regimens for malaria

Regimen Dose for adults Dose adjustment for renal function

Mefloquine 250 mg weekly No Doxycycline 100 mg daily No Atovaquone/proguanil 1 tablet daily Do not use if GFR <30 mL/min Proguanil plus chloroquine 200 mg daily plus Yes for proguanil 300 mg (base) weekly Chloroquine 300 mg (base) weekly No Proguanil 200 mg daily Yes Travel vaccinations 261 people with other medical conditions, including • Ferrous sulfate may reduce the serum levels renal failure. of doxycycline by up to 90%. • Chloroquine absorption may be reduced by calcium carbonate. Additional factors to consider for renal • Calcium- and aluminium-based phosphate patients binders may reduce doxycycline levels by 50–100%. Dose adjustment Proguanil is excreted by the kidney and there- Specialist advice fore requires dose adjustment according to When taking into account all the above factors renal function (Table 20.3).5 Patients with renal it may prove difficult to decide which, if any, is failure receiving proguanil should also be pre- the most appropriate antimalarial for a particu- scribed folic acid 5 mg daily to reduce haema- lar patient. In such cases detailed advice is avail- tological side-effects.5 able from the Malaria Reference Laboratory (at Malarone (atovaquone 250 mg/proguanil the London School of Hygiene and Tropical 100 mg) is not recommended at GFR <30 mL/ Medicine) or the National Travel Health min. The dose of proguanil would need to be Network and Centre. All options should also be reduced, but full dose of atovaquone would be discussed with the renal clinician. It may be required. This is clearly not possible with a that the advice is not to travel to a particular combined product. No dose adjustments are area. required for any of the other agents when used at prophylactic doses.

Travel vaccinations Interactions Renal patients often take multiple medications. The requirement for travel vaccinations will be It is important to check carefully for possible determined by the disease risk to the traveller. interactions when selecting the most appropri- All travellers should undergo a full risk assess- ate antimalarial agent. Interactions could result ment before travel. Travel-related hazards and in antimalarial effects being reduced or blood risks may be determined not only by the coun- levels of immunosuppressants being altered. For try or countries to be visited, but also the area example:12 to be visited within that country (city or rural, • Ciclosporin, tacrolimus and sirolimus plasma altitude or jungle, etc.), type of accommodation, concentration levels can be increased by both and proposed activities to be undertaken. Some doxycycline and chloroquine. risks may also be seasonal. It is vital, therefore, that guidance is given based on current recom- mendations. The National Travel Health Network and Centre (NaTHNaC) telephone advice line provides guidance for health profes- Table 20.3 Dose of proguanil in renal sionals advising travellers who have a complex impairment medical history or travel itinerary or both. GFR (mL/min) Prophylactic dose of proguanil Information about travel-related health issues, for adults including clinical updates and disease out- breaks, can be found at the NaTHNaC website >60 200 mg daily (www.nathnac.org). The on-line Travax database 20–59 100 mg daily (www.travax.scot.nhs.uk – requires subscription) 10–19 50 mg alternate days provides up-to-date country-by-country advice <10 50 mg once a week regarding travel vaccinations, malaria, disease CAPD/haemodialysis 50 mg once a week outbreak news and other travel health-related 262 Chapter 20 • Travelling and vaccines issues. Fit for Travel (www.fitfortravel.scot.nhs. uk) is provided by NHS Scotland and is intended Table 20.4 Vaccines given for travel when as a travel health resource for the general pub- indicated (doses may differ for children and lic. All the websites have links to other valuable adults) travel health-related websites. Renal patients have special immunisation Inactivated vaccines Live attenuated vaccines requirements and contraindications, so it is Hepatitis A Yellow fever important that the traveller provides full med- Hepatitis B BCG ical details as well as a detailed travel itinerary Typhoid Typhoid (oral) when accessing travel advice. The advising Oral cholera MMR health professional should confirm that a trav- Influenza Varicella (rarely used in UK) eller’s primary courses and boosters have been Pneumococcal received as recommended in the full British Polio schedule. In addition, the Department of Health Diphtheria, polio and recommends that patients with chronic kidney tetanus (combined) disease receive influenza and pneumococcal Japanese encephalitis immunisation. All patients being considered for, Meningococcal or who are on, dialysis should also be vaccin- meningitis – ACW135Y ated against hepatitis B. Rabies Immunosuppressed patients, including both Tick-borne encephalitis transplant patients and those on dialysis, should Haemophilus influenzae not receive live vaccines. Inactivated vaccines type B can be administered, although the response may be reduced. Patients with end stage renal disease countries where the disease is endemic.13 Many (ESRD) have a reduced response to vaccination countries, although not the UK, require an because of the general suppression of the International Certificate of Vaccination or Pro- immune system associated with uraemia. The phylaxis from travellers arriving from, or who response of transplant patients to vaccination have passed through, an endemic area, before will depend on their degree of immunosuppres- entry is allowed. However, countries for which sion and underlying disease. See Table 20.4 for the vaccine is recommended may not them- live/inactivated vaccines. selves request a vaccination certificate, so the absence of a requirement for a vaccination certificate does not imply that there is no risk in Hepatitis B that country. Yellow fever is a live vaccine, and as such, The hepatitis B status of a haemodialysis patient should not be given to immunosuppressed (in addition to other blood-borne viruses such travellers. Renal patients who are immuno- as hepatitis C and HIV) may be requested by the suppressed should avoid travel to yellow fever holiday dialysis centre within a certain number endemic areas as they cannot be protected of weeks before travel, and boosters may be against the disease by vaccination. A letter of required. Each haemodialysis unit in the UK will medical exemption may be provided if an indi- have a policy for the isolation of patients vidual should not be vaccinated on medical returning from dialysis in high-risk areas outside grounds, and is visiting low-risk areas where a Europe and North America. certificate requirement exists. Yellow fever vaccine can only be adminis- tered at designated yellow fever centres. Yellow fever Designated centres in England and Wales are listed on the National Travel Health Network Yellow fever vaccination is recommended for and Centre website (www.nathnac.org). In- travel to some African and South American formation on country requirements for yellow Conclusion 263 fever is published annually by the World Health may be that the patient should not travel to a Organization in International Travel and particular destination. Health, Vaccination Requirements and Health Advice (www.who.int). International Certific- ates of Vaccination or Prophylaxis are valid Conclusion for 10 years starting 10 days after the date of vaccination. This chapter has given a brief overview of some of the considerations for which travellers from Vaccination summary the UK with renal disease may seek advice. Although there are some restrictions, in the Although not always possible, travellers should main, patients with renal disease are able to plan vaccinations at least eight weeks ahead of travel to many areas of the world, provided they travel, to ensure that the recommended time make appropriate arrangements, take the interval between doses and vaccines is followed. required medications, and follow closely This will also allow immunity to develop before the travel health advice they are given. departure. It may be necessary to discuss immunisation requirements for a renal patient with the super- Acknowledgements vising clinician in addition to a specialist centre such as NaTHNaC. The advice, based on vac- With thanks to Hilary Simons, Claire Stringer, cination requirements and contraindications, and Dr Lisa Ford for their comments and advice.

C CASE STUDIES

These cases are given as examples only. Always use up to date reference sources when advis- ing actual patients. Seek expert medical opinion for complex patients and/or complicated travel plans.

Case 1 Ellen is a 43-year-old English woman who plans a 10-day cruise which will incorporate travel to the USA, Mexico, Panama Canal, Colombia (Cartegena only), and Barbados. She has chronic renal failure secondary to systemic lupus erythematosus, but does not yet require dialysis. Her current medication includes prednisolone.

Q1. What are the vaccinations required for travel to these areas?

Q2. What other considerations need to be taken into account when recommending appropriate vaccinations for Ellen?

Q3. What malaria prophylaxis is appropriate?

(continued overleaf) 264 Chapter 20 • Travelling and vaccines

C CASE STUDIES (continued)

Case 2 Dorothy is a 56-year-old woman with renal failure secondary to diabetes, now requiring haemodialysis. She is planning a three-week trip through South Africa (Cape Town, Durban, Kruger Park) and onward overland to Zambia (Victoria Falls), returning to South Africa via the Chobe National Park, Botswana. Her current medications are: ramipril, amlodipine, calcium carbonate, alfacalcidol, doxazosin, furosemide and insulin.

Q1. What would be appropriate malaria prophylaxis for travel to these areas?

Q2. Which would be the most appropriate chemoprophylaxis for Dorothy?

Q3. What advice about malaria prophylaxis should be given to Dorothy?

Q4. Which vaccinations are required for travel to these areas?

Case 3 Mary plans a two-week all inclusive hotel holiday to Punta Cana in the Dominican Republic. She had a renal transplant in 1997 and is taking ciclosporin. Her GFR is now 55 mL/min. She is cur- rently well, although she has been treated in the past for post-natal depression. On a previous trip 2 years ago, during which she took proguanil 200 mg daily, there was significant disturb- ance to her renal function.

Q1. What would be appropriate malarial prophylaxis for travel to Punta Cana?

Q2. Which agent would be most appropriate for Mary?

Q3. Would any other antimalarial be suitable?

Q4. What advice would you give to Mary?

References April 2005. www.nathnac.org/pro/factsheets/ food.htm (accessed 16 April 2006). 5. Ashley C, Currie A, eds. The Renal Drug Handbook, 1. Bordea C, Wojnarowska F. Educating renal trans- 2nd edn. Oxford: Radcliffe Medical Press, 2004. plant patients about skin cancer. Br J Ren Med 6. NaTHNaC Travel Health Information Sheet, 2003; 8: 23–26. Insect Bite Avoidance April 2005. www.nathnac. 2. Paterson P, Hart S, Cahill S. What we tell our org/pro/factsheets/iba.htm (accessed 16 April patients about OTC medicines. Br J Ren Med 1999; 2006). 4: 13–16. 7. British Medical Association Board of Science and 3. Mason P. What advice can pharmacists offer trav- Education. The impact of flying on passenger ellers to reduce their health risks? Pharm J 2004; health: a guide for healthcare professionals. May 273: 651–656. 2004. www.bma.org.uk/ap.nsf/Content/Flying? 4. NaTHNaC Travel Health Information Sheet, OpenDocument&Highlight=2,impact,flying (ac- Prevention of Food and Water Borne Diseases cessed 16 April 2006). Sources of further information 265

8. Goodyear L. Medical kits for travellers. Pharm J Health – covers vaccination requirements and 2001; 267: 154–158. travel health advice. ISBN: 9241580275. Annual 9. Chiodini P, Hill D, Lalloo D et al. Guidelines publication available online at: www.who.int/ith/ for malaria prevention in travellers from the index.html United Kingdom. 2006. http://www.hpa.org.uk/ Department of Health. Immunisation against infectious publications/2006/Malaria/Malaria_guidelines.pdf disease. The Green Book. London: HMSO 2006. Also 10. Hughes C, Tucker R, Bannister B, Bradley DJ. Mal- available online at www.dh.gov.uk/PolicyAnd aria prophylaxis for long-term travellers. Commun Guidance/HealthAndSocialCareTopics/fs/en Dis Public Health 2003; 6: 200–208. Department of Health. Health Information for Overseas 11. Department of Health. Health Information for Travel. The Yellow Book. London: HMSO, 2001. Avail- Overseas Travel. The Yellow Book. London: HMSO, able at www.nathnac.org (Due for update soon) 2001. 12. Baxter K, ed. Stockley’s Drug Interactions, 7th edn. London: Pharmaceutical Press, 2006. Electronic information 13. NaTHNaC Travel Health Information Sheet, Yellow Fever August 2005. www.nathnac.org/pro/ British Foreign Office travel warnings: www.fco.gov.uk factsheets/yellow.htm (accessed 16 April 2006). NHS Scotland Fit for Travel website: www.fitfortravel. scot.nhs.uk Health advice for travellers leaflet available free from Further information Post Offices and online: www.dh.gov.uk/travellers National Kidney Federation holiday pages: www. kidney.org.uk Guidelines for the prevention of malaria in travellers National Travel Health Network and Centre from the United Kingdom. Commun Dis Public (NaTHNaC): www.nathnac.org Health 2003; 6: 180–199. Available online at www. Travax travel database (requires subscription): www. hpa.org.uk/infections/topics_az/malaria/menu.htm travax.scot.nhs.uk World Health Organization: International Travel and World Health Organization: www.who.int

Case study answers

Chapter 3 renal failure. Just provide supportive meas- ures, treat the symptoms and, if possible, the Q1. What patient and pharmaceutical factors underlying cause, and wait to see if renal may have precipitated acute renal failure in function is recovered. this patient? • Stop all oral medications except the raniti- • The patient is elderly. dine – the patient is at risk of developing • He has previously been prescribed bendro- stress ulceration. flumethiazide on an escalating dose. This in • Treat the hyperkalaemia – this is the most turn precipitated an episode of gout. At this life-threatening symptom at present. point the thiazide should have been stopped, Initially, the patient could be given salbut- since thiazide diuretics are contraindicated in amol nebs, followed by insulin and glucose gout. This did not happen. (± calcium gluconate IV), then, if necessary, • The colchicine prescribed to treat the gout Calcium Resonium and lactulose. caused the patient to vomit repeatedly, thus • Blood glucose levels should be very closely dehydrating him. monitored, especially considering the treat- • The GP then added in indometacin, which ment of the hyperkalaemia, and the switch further compromised renal perfusion. The from gliclazide to insulin. combination therapy of an NSAID, an ACE • Although the patient is usually hypertensive, inhibitor and a diuretic in an already dehy- the fact that he is currently severely dehy- drated patient induced a state of acute renal drated would tend towards a state of failure. hypotension. Therefore, all antihypertensive • A previous medical history of non-insulin- medications should be stopped temporarily dependent diabetes mellitus (NIDDM) and until the patient is euvolaemic, and then hypertension are added risk factors for devel- reintroduced as necessary. oping acute renal failure, as they will predis- • Treat the metabolic acidosis with sodium pose the patient to having a degree of bicarbonate, either IV or PO. chronically impaired renal function. • The hyperglycaemia will respond to the • Acute gout could cause urate uropathy, insulin and glucose used to treat the hyper- whereby uric acid crystals are deposited kalaemia. The patient could then be put on within the renal tubules, causing intrinsic a sliding scale insulin infusion, until such renal damage. time that he is considered stable enough to reintroduce the gliclazide. • Since the patient is dehydrated, he should Q2. What are the main pharmaceutical receive crystalloid by IV infusion until problems and how might they be managed? euvolaemic. Ideally, his CVP should be mon- • Main pharmaceutical problems are acute itored in order to assess fluid balance. Once renal failure, hyperkalaemia, metabolic aci- the patient is rehydrated, if he still fails to dosis, hyperglycaemia and dehydration. pass urine, stop giving large volumes of fluid • There is no actual treatment per se for acute replacement. To continue to administer large

267 268 Case study answers

volumes of fluid would put the patient at risk 1.23 (140 – Age) Weight of developing severe hypertension and pul- CrCl = Serum creatinine monary oedema. Furosemide should only be = 1.23 (140 – 65) 68 given if the patient is euvolaemic or fluid 272 overloaded, and if the drug fails to induce a diuresis, then it should be discontinued, as i.e. CrCl = approx 23 mL/min. further use merely increases renal damage. At this level of renal function, a dose of There is no evidence that either mannitol or gentamicin 3–4 mg/kg once every 24 hours dopamine are of any benefit. could be used, and trough levels monitored, to ensure they are below 1.5–2.0 mg/L before the next dose is given. Q3. Comment on the appropriateness of the prescribed antibiotic therapy. What advice would you provide regarding this? Mr VC has recovering renal function after an epi- Chapter 4 sode of acute renal failure, but at this stage any futher nephrotoxic insults should be avoided. Q1. What are the metabolic abnormalities? Ideally, Mr VC should have been prescribed • Renal impairment – acute on chronic prophylactic antibiotics to cover the insertion of • Hyperkalaemia – life threatening the catheter. Suitable agents would include • Low bicarbonate – from renal impairment ciprofloxacin 500 mg PO, or if gentamicin was causing metabolic acidosis warranted, a single dose of 80 mg would have • Hyperphosphataemia been sufficient. • Anaemia. Since Mr VC developed urinary sepsis, even without benefit of cultures and sensitivities, it would be reasonably safe to assume that the Q2. What has caused the abnormalities? patient has a Gram-negative sepsis. As such, oral • Angiotensin II receptor blocker – most likely. (500 mg twice daily) or IV (200 mg twice daily) The history of ischaemic heart disease and ciprofloxacin would be a good choice. An peripheral vascular disease with hypertension aminoglycoside would not be first-choice ther- suggests renovascular disease. apy since the associated nephrotoxicity would • Could possibly have any other cause (i.e. vol- damage the recovering kidneys, and the accu- ume depletion, obstruction, sepsis, etc.) but mulated aminoglycoside also puts the patient at this is unlikely given the patient’s history. increased risk of ototoxicity. However, if it were deemed to be necessary, the dose should be adjusted according to the patient’s renal Learning points function. Some centres may wish to use an alternative, • ACE/angiotensin II blockers are not contra- less nephrotoxic antibiotic, for example, a indicated in renal impairment. They are the second- or third-generation cephalosporin IV, only antihypertensive agents to reduce the co-amoxiclav, ciprofloxacin, etc. Trimethoprim progression of renal disease significantly in will probably not be potent enough to treat full- proteinuric patients. All renal failure patients blown urinary sepsis. In addition, anaerobic should be on them – especially those with cover may be required, for example, with diabetes, the commonest cause of ESRF. In metronidazole. addition, the majority of ESRF patients die of Since Mr VC has fairly rapidly changing renal a cardiovascular event, and many on dialysis function, the Cockcroft and Gault equation develop left ventricular hypertrophy, often should be used with great caution. However, as secondary to chronic fluid overload. ACE a rough calculation of his current degree of inhibitors are the drug of choice to aid renal impairment: left ventricular remodelling and decrease left Case study answers 269

ventricular hypertrophy. Renal artery stenosis way of clearing potassium (e.g. 500 mg to 1 g is the only renal contraindication to their per day). use. Mild hyperkalaemia is relative and usu- • Calcium Resonium – works well but compli- ally controllable. ance is a problem. Needs to be administered • Angiotensin II blockers, like ACE inhibitors, with lactulose to stop bowel congestion. are contraindicated in renovascular disease. Takes a few days to work • Angiotensin II blockers do cause hyper- • Dialysis/haemofiltration – in uncontrollable kalaemia but perhaps to a lesser extent than potassium – is the only way to rapidly ACE inhibitors. remove potassium. Has associated risks for • The combination of ACE inhibitors/ patient on dialysis. angiotensin II blockers gives synergistic reduction in blood pressure and proteinuria Q4. What is the rest of his medication for and but also increases the risk of hyperkalaemia. how much does it cost per year? • Most patients will show some mild deteriora- tion in function after starting ACE/ • Calcium carbonate – primarily used as a angiotensin II blocker but do not necessarily phosphate binder to limit the amount of have renal artery stenosis dietary phosphate absorbed from the gut. Any excess calcium can be absorbed to treat the hypocalcaemia associated with ESRF. Q3. What is the life-threatening complication Other phosphate binders commonly used and how would you treat it? are: calcium acetate, magnesium carbonate, Hyperkalaemia. aluminium hydroxide, sevelamer hydro- chloride. • Alfacalcidol – for control of PTH aim for 2–3 Learning points times upper limit of normal. Vitamin D is a • Calcium gluconate – to stabilise myocardium steroid hormone, and there are vitamin D – competition between potassium and cal- receptors on the parathyroid glands which cium for cardiac muscle binding sites. help regulate the production of PTH, and Calcium gluconate does not reduce potas- hence control bone turnover. ESRF is associ- sium and is very venotoxic. Should be given ated with decreased active vitamin D levels, into a large vein. which coupled with hypocalcaemia and • Insulin and dextrose – shifts potassium into hyperphosphataemia, eventually leads to cells. Insulin receptor has a potassium the development of secondary hyperpara- channel associated with it. This is very effec- thyroidism and hence to renal bone disease. tive at reducing plasma levels of potassium, • Amlodipine – high blood pressure affects but does not get rid of potassium. It is only almost all chronic renal failure patients. a temporary measure which lasts a few hours Ischaemic heart disease is much higher than before potassium levels will begin to rise in general population (50–500 times risk). again. Patients must receive close monitoring Risk of a 35 year old on dialysis dying of of blood glucose levels. ischaemic heart disease is the same as that of • Salbutamol (nebulised or IV) – effective, espe- an 85 year old in the general population. cially if patients are bradycardic with hyper- • Atorvastatin – see above. Lipid profiles are kalaemia. There is a potassium channel different in renal failure. Lower total choles- associated with the beta receptor. terol but adverse LDL:HDL ratios. Also higher • Lactulose +++ – diarrhoea is a very effective triglycerides. way to reduce excess fluid and potassium in • Vitamin supplementation (Ketovite) – ESRF the body. Beware if already intravascular patients are notorious for having poor volume deplete as it will cause profound nutrition. Hyperkalaemia can be life- hypotension. threatening, so fruit, vegetables and foods • Diuretics – high-dose furosemide is a good generally containing water-soluble vitamins 270 Case study answers

are on a restricted diet list, therefore patients • It could be that she has had an acute bleed – may require supplements. There is a differ- she has CKD, is on an NSAID, and has no ence between the tablet and liquid forms. gastro-protection. Has the fall in haemo- Tablets are water-soluble vitamins, whereas globin been acute or chronic? liquid contains fat-soluble vitamins – you cannot interchange them. • Iron sulfate – due to increased red cell Q2. If ESA therapy is to be initiated, what are the choices in terms of initial dose and turnover, reduced absorption, increased need monitoring? with erythropoietin (EPO) therapy. Assess- ment of iron status in CRF patients is notori- Since Mrs AP is not on haemodialysis, she needs ously difficult. There is evidence of altered her therapy administered by the subcutaneous gut pH in renal patients, which coupled with route, and at the time of preparation this meant gut oedema from fluid overload makes renal therapy with either NeoRecormon (epoetin patients very poor absorbers of oral iron. beta) or Aranesp (darbepoetin). NeoRecormon • Furosemide – for control of fluid status and could be started at 60 units/kg weekly – if Mrs salt load. Doses of 500 mg to 1 g daily are AP were 60 kg this would be 3600 units weekly. not uncommon. Still works even if GFR Most centres would prescribe this at 2000 units <5 mL/min. twice a week, to reduce the number of injections • Sodium bicarbonate – to reduce acidosis and that Mrs AP needs, though three times a week therefore reduce protein breakdown and would be more efficient. Haemoglobin should bone buffering of H+ ion. It may lead to a rise at 1–2 g/dL monthly, and if this is not huge salt load (8.4% sodium bicarbonate = happening most prescribers would increase to 1 mmol/mL therefore 1 L will give a load of 4000 units twice weekly, and eventually to 1000 mmol Na+. One 600 mg tablet contains 6000 units twice a week. Blood pressure, full approx. 7 mmol bicarbonate). May help with blood count, iron stores and potassium need hyperkalaemia, but only in minor way. monitoring. If Aranesp is used, the initial dose • EPO – target haemoglobin is approximately of 0.45 µg/kg weekly suggests a prescribed dose 11 g/dL. of 30 µg weekly, monitored and adjusted in a • Cost is approximately £4000–6000 per similar manner. annum.

Q3. What factors might contribute to reduced Chapter 5 efficiency of ESA therapy in Mrs AP? The above answer assumes that her iron stores Q1. The Senior House Officer (SHO) asks if Mrs are adequate to ensure erythropoiesis, and if not AP could be started on ESA therapy. Give three have been corrected with intravenous iron ther- reasons why you might advise against this at apy. In a non-haemodialysis patient, oral iron this point. supplementation with ferrous sulfate may • Her blood pressure is currently uncontrolled maintain adequate transferrin saturation during – it would make sense to control this before erythropoiesis. Care should be taken to avoid initiating a drug that can elevate blood pres- interactions between phosphate binders and sure further. iron salts. In addition, Mrs AP has a degree • No iron studies have been performed at this of uraemia and hyperphosphataemia, both of point. It could be that she is iron depleted which impair erythropoiesis, and factors such as

and this is the cause of her anaemia. At this her PTH, folate, B12 and so forth are unknown. level of renal impairment, it may be that If angiontensin-converting enzyme (ACE) erythropoietin deficiency is more likely, but inhibitors were initiated for her hypertension, unless any iron deficiency is corrected then these can affect erythropoiesis as well, and ESA therapy will not be effective in raising finally aluminium from her AluCap capsules haemoglobin. may be having an adverse effect. Infections and Case study answers 271 inflammation can reduce response to ESAs cially in the case of Mrs A because she has co- dramatically. existing hypocalcaemia. For example, calcium carbonate (Calcichew) at a dose of one or two Q4. What factors affect the choice of ESA for tablets 5–10 minutes pre-meals may be pre- Mrs AP? scribed. Patients need to have some knowledge of why a high phosphate is important (e.g. con- All the companies have evidence that their prod- tributes to bone damage), how the drugs work uct offers certain advantages in different situ- by binding dietary phosphate in the gut (so are ations, but in general, both Aranesp and very different from their other medicines as NeoRecormon achieve similar responses to ther- they are taken at mealtimes only and dose may apy, have similar rates of adverse drug reactions vary according to type of meal) and the most and cost about the same (depending on local common binder side-effects (e.g. gastrointest- contracts). Aranesp offers reduced dosing fre- inal disturbances). quency without loss of efficiency, which means fewer injections for non-haemodialysis patients. Q4. What do you think is a realistic target NeoRecormon has a longer track record, a range serum phosphate level for Mrs A? of different administration devices, and a very fine needle on the pre-filled syringe and pen It is generally accepted that achieving a normal device which patients report favourably upon. phosphate level is unlikely for the majority of With the rise of the regional contract in the UK, CKD patients. Realistic targets in current guide- the choice of product is increasingly being lines are serum phosphate less than 1.7 or removed from individual prescribers. 1.8 mmol/L. Patients should be aware of what their target phosphate is.

Chapter 6 Q5. Is vitamin D therapy indicated for Mrs A at this point? If yes, which vitamin D agent and Q1. Why is this patient’s hyperphosphataemia what starting dose would you recommend? If worsening? no, why do you think it is appropriate to leave The progression of CKD further impairs the her on Calcichew alone at present? urinary excretion of phosphate. Compensatory Ideally, Mrs A would not be commenced on mechanisms to promote phosphate clearance vitamin D at this stage. Her mild hypocalcaemia can no longer cope as renal function declines could be corrected by Calcichew alone, through and serum phosphate will continue to rise. gastrointestinal absorption of calcium and reduction in serum phosphate, which will Q2. Are there any symptoms she may now increase the proportion of free serum calcium. experience secondary to her elevated In practice, some patients would be commenced phosphate? on vitamin D at this stage – alfacalcidol 0.25 µg Hyperphosphataemia contributes to uraemic daily is a commonly prescribed regimen. itching and can also cause redness and soreness in the eyes. Q6. What other biochemical investigation(s) would you request at this stage in the management of Mrs A’s renal bone disease? Q3. What would you advise as first-line How would knowledge of the result(s) phosphate-binding agent for Mrs A? What dose influence your decision to initiate or hold off would you recommend? How would you vitamin D therapy? educate the patient on why phosphate binders are important, how they work, when to take A serum parathyroid hormone (PTH) is indi- them and what side-effects to expect? cated for Mrs A. It is a vital part of the bio- Calcium-based phosphate binders are currently chemical bone profile used to guide prescribing the first-line agents in many CKD patients, espe- of phosphate binders and particularly vitamin 272 Case study answers

D. A high PTH would indicate a significant patients can lead to excessive suppression of degree of secondary HPT is already present – PTH secretion, which increases the risk of induc- requiring prescription of vitamin D for its direct ing low bone turnover forms of renal osteo- and indirect effects to suppress PTH secretion. dystrophy (e.g. adynamic bone disease). Target In CKD patients, vitamin D is used mainly for levels for PTH in CKD are not normal range – its actions to control secondary HPT, not simply for example in ESRD usually aim for 3–5 times correcting hypocalcaemia. A mildly raised PTH upper limit of normal range. would mean less need for vitamin D at present – controlling phosphate alone will directly sup- Q10. What would you suggest as a vitamin D press PTH secretion. plan in Mrs A and what are the reasons behind your decision? Stop alfacalcidol? Reduce Q7. What are the other two commonly alfacalcidol dose? Continue alfacalcidol at prescribed phosphate binders available to the current dose? Increase alfacalcidol dose? nephrology team? Using the serum PTH alone, Mrs A needs a dose Aluminium hydroxide (AluCap) or sevelamer increase in her alfacalcidol but the elevated hydrochloride (Renagel). calcium and (to a lesser extent) phosphate effec- tively contraindicate this plan at the moment. Q8. Considering the advantages and However, the hypercalcaemia is only mild so the disadvantages of each of these agents, which benefits of continuing alfacalcidol to continue one would you recommend and what would be management of HPT probably outweigh the your suggested starting dose? risks of stopping it and seeing PTH rise even Aluminium less likely to be used by nephro- higher. Reducing the dose (e.g. to 0.25 µg daily) logists these days. Concerns over toxicities such or even continuing current dose appear to be as dementia, anaemia and adynamic bone dis- reasonable options, particularly as you have ease, though controversial, mean that many now switched Mrs A to a calcium-free phos- renal units only use it short term, particularly as phate binder. In either case, it is very important an additional agent to control severe hyper- to check the bone biochemistry more frequently phosphataemia. However, aluminium binders during this period of hypercalcaemia, for exam- are cheap and potent – a dose of one AluCap ple in two weeks time, to review response to capsule with meals would be an effective start- changes in the renal bone disease drug regimen. ing dose for Mrs A. Sevelamer would be a more likely prescribed drug in many renal units. It has Q11. What form of renal osteodystrophy is the benefit of being calcium- and aluminium- likely to have contributed to Mrs A’s fracture free but is expensive and has gastrointestinal and how does it influence bone turnover? side-effects to match other binders. Also, it is less Mrs A’s bone disease predominantly appears potent than aluminium- and probably calcium- related to secondary HPT, in view of the raised based binders so in this case Mrs A would need PTH and alkaline phosphatase. This is known as to start on two or three tablets with meals. osteitis fibrosa. The excess PTH is acting on the bone to increase activity of both osteoblasts and Q9. How can vitamin D therapy cause especially osteoclasts so it is classed as a high complications in the management of renal osteodystrophy? bone turnover form of renal osteodystrophy. The effectiveness of vitamin D in management Q12. Why are the doctors concerned about of secondary HPT can be limited by its dose- extraskeletal manifestations of an abnormal related actions: bone biochemistry profile (i.e. calcification) in • Hypercalcaemia Mrs A? At which sites in the body can • Exacerbation of hyperphosphataemia. calcification occur in a CKD patient? How can calcification impact on the morbidity and Also, overzealous use of vitamin D in CKD mortality of CKD patients? Case study answers 273

Calcification is increasingly recognised as a sig- (from the Sheffield table14) and indicates a risk nificant problem in CKD patients and can be of CHD >30% over 10 years. Treatment with a present at various sites, for example: statin and aspirin is indicated. Even without the Sheffield table, diabetes is a risk factor for CHD, • Soft tissue and peripheral vasculature (in its as is hypertension and a cholesterol >5 mmol/L. most severe form this can progress to calci- Therefore a statin should be started, at low dose phylaxis) considering his renal impairment. • Skin, joints, lungs, cornea, conjunctivae and muscle • Cardiovascular system (myocardium, coron- Q2. What treatment would be suggested with ary arteries, aorta and cardiac valves). respect to his blood pressure? Calciphylaxis is rare but difficult to manage He has been hypertensive for some time so drug and is associated with a high mortality rate. therapy is indicated. Considering his diabetes Cardiovascular calcification is a much greater an ACE inhibitor would be first choice, with a problem because the most common causes of calcium channel blocker second line. Monitor morbidity and mortality in CKD patients have renal function closely. cardiovascular aetiology. Cardiovascular disease in CKD patients is multifactorial in origin but Q3. What other advice would be appropriate? evidence is accumulating for the importance of the role of calcification and how disturbances To minimise his CHD risk, he should be advised of bone biochemistry influence this. to stop smoking, given advice on a healthy, low- salt diet, and encouraged to exercise regularly Q13. What are the benefits of prescribing and keep alcohol intake low. He should be higher doses of calcium-free phosphate binders referred to a nephrologist and pre-dialysis clinic (sevelamer and/or aluminium hydroxide) for to minimise his renal disease. Mrs A in relation to preventing further bone fractures and reducing her calcification risk? For Mrs A, knowledge of the risks of calcifica- Chapter 8 tion mean that her abnormal bone biochemistry Q1. At the monthly multidisciplinary meeting, needs to be managed more aggressively and not a haemodialysis patient’s urea reduction ratio is simply to protect her bones. Increasing phos- only 50%. What can be done to improve their phate binder doses will hopefully bring down adequacy? both serum phosphate and PTH which will have benefits for the bones. More intensive use of Adequacy can be improved by increasing the calcium-free phosphate binders in Mrs A may size of the dialyser, time on dialysis, increasing also reduce her calcification risk and potentially the blood flow (the patient’s access may restrict provide an associated reduction in cardio- this). If there are problems with access then the vascular risk through the following actions: patient should be referred to the vascular sur- geon. If possible the patient could be changed • Reducing hyperphosphataemia to haemodiafiltration. • Reducing calcium burden by avoiding calcium-based phosphate binders Q2. Miss RB is an 80-year-old woman recently • Correcting severe HPT with associated fall in started on haemodialysis. After about an hour serum calcium on dialysis she becomes hypotensive. What can • Reducing calcium phosphate product. be done about it? The first thing would be to get Miss RB’s dry Chapter 7 weight assessed. In the acute situation, fluid should be given to the patient and her feet Q1. What risk factors does he have for CHD? raised above her head. In the longer term, anti- A TC:HDL-C ratio of 6.9 exceeds the ratio of 6.0 hypertensive medication could be omitted on 274 Case study answers dialysis days or given post dialysis. If she is constipation. If constipation is confirmed then not on any blood pressure medication then treatment with laxatives should be commenced, midodrine (unlicensed) given 1 hour before if there is no success with senna and lactulose dialysis or fludrocortisone may be commenced. then stronger laxatives such as Picolax may be Sometimes stopping the patient from eating on required. dialysis can help. In an ideal world, changing to haemofiltration could help, but that is rarely Q5. On checking the notes it appears that Mr practical. If possible, the patient could be BA has had a couple of peritonitis episodes in changed to haemodiafiltration, which can the past few months and is a known staph improve cardiovascular instability. Again, that is carrier. What would you recommend? not available in every unit. He could be started on prophylactic nasal mupirocin for 5 days a month in case that is the Q3. Mr MP has recently started on CAPD and reason for his repeated episodes of infection. has been having problems with pain on The peritoneal dialysis team should also check draining in his fluid. What could be the reason for his pain and what dialysis fluid should he his technique in case he is having problems at use? home. The reason for Mr MP’s pain could be that the dialysis fluid he is using is too acidic for his peri- Q6. Miss RB did not have good enough veins toneal membrane and that is causing him pain. for a fistula so has a permanent catheter in place. She has come in today and the line is not It could also be that too much fluid is being working. What can be done? drained in and he may need the volume reduced initially. Mr MP could also be changed Both urokinase and alteplase can be used to to a more biocompatible fluid with bicarbonate unblock lines. A lock of 5000 IU urokinase or added to it (e.g. Physioneal or Balance). Before 2 mg alteplase may be tried initially. Both would these fluids were available sodium bicarbonate be left in for an hour. If this did not work then or lidocaine were added to the bags, but that an infusion of urokinase 10 000–250 000 IU sometimes leads to an increased incidence of over 3–24 hours or alteplase 20–50 mg over peritonitis as aseptic technique cannot always 12–20 hours would be done. If all else fails, the be assured. line would have to be removed. If it became a chronic problem then she might be started on low-dose aspirin or clopidogrel if she did not Q4. Mr BA is a 45 year-old man on peritoneal dialysis. He was admitted with abdominal pain tolerate the aspirin. the previous evening. What are the possible causes of his abdominal pain and what should Q7. Mr MC has only been on haemodialysis a be done? few weeks and it was noticed that his platelet Possible causes of abdominal pain are constipa- count has fallen quite dramatically. Heparin- tion or peritonitis. A sample of the peritoneal induced thrombocytopenia (HIT) has been diagnosed. What can be used as an fluid should be sent off for white cell count and anticoagulant for his dialysis? cultures and sensitivities. If the bag is cloudy then peritonitis should be assumed and anti- In patients with HIT one of the first things to biotics started empirically (e.g. vancomycin and try would be heparin-free dialysis. If anticoagu- gentamicin) and altered once sensitivities are lation was required then epoprostenol could be known (antibiotics may vary between different used, although it can be quite difficult to get the units). His white cell count came back at 1926 correct flow rate. Other alternatives would be a mm3 with 80% polymorphs. The organism iso- bolus dose of danaparoid 2000–3750 units or lated was Staph. epidermidis, which is sensitive to lepirudin 0.14–0.15 mg/kg pre-dialysis or a vancomycin, so the gentamicin was stopped. An bolus dose and continuous infusion of argatro- abdominal X-ray should be taken to check for ban 2–3 µg/kg/min (started at least 4 hours Case study answers 275 before dialysis) or as a bolus of 250 µg/kg at the bleeding risk has diminished. There is an endur- start of dialysis followed by a continuous infu- ing indication for a statin. Simvastatin could be sion of 2 µg/kg/min. continued as tacrolimus is not known to signi- ficantly increase the risk of myopathy; however, some units would opt to use a lower dose or an Chapter 9 alternative statin. Subcutaneous insulin would usually be Q1. What recipient factors might have led to JL replaced with an intravenous sliding scale peri- remaining on the waiting list for over 3 years? operatively. Average waiting times for a kidney transplant are in the order of 2 years. The renal transplant Q3. What other antirejection strategy might allocation system is complicated and takes into have been considered and could it be justified? consideration ABO blood group, HLA tissue- Induction immunosuppression with an IL-2R type matching, known and declared recipient antibody such as basiliximab is an option which HLA or ABO antibodies, time on the waiting list is supported by NICE in combination with a and the age and size of both the donor and tacrolimus-based triple therapy. In this case the recipient. patient has a poorly HLA-matched graft which JL has waited longer than average. It is pos- is a risk factor for acute rejection and so this sible that she would have anti-HLA or ABO anti- strategy would be justified. bodies from her pregnancies or blood transfusion which might have resulted in a longer wait for a compatible match. It is also Q4. Is this tacrolimus level usually considered possible that she might have been suspended adequate? from the transplant waiting list for the periods Early tacrolimus levels of between 10 and when she had active peritonitis. 15 ng/mL are more normal though some centres opt to use lower doses, particularly where there Q2. Which medicines should be stopped at this is a high risk of delayed graft function. point and which would normally continue to be prescribed? Q5. What might be contributing to this level? The epoetin, B-group vitamins, folic acid, phos- There is great inter-patient variability in the phate binder and alfacalcidol can all safely be dose of tacrolimus required to achieve target stopped once a patient is transplanted, even if trough levels. This is the main justification for there is delayed graft function. Alfacalcidol is intensive early therapeutic drug monitoring. It usually continued only if the recipient has had is also possible that the carbamazepine required a parathyroidectomy. for her diabetic neuropathy has induced higher It is usual to stop antihypertensive medica- activity in liver and gut wall cytochrome P450 tions and review if there is a continued need isoenzymes resulting in greater tacrolimus clear- once the kidney is working. Most units will ance and lower than expected trough levels. delay starting an ACE inhibitor in the early post-operative period as sudden increases in serum creatinine can be difficult to interpret Q6. What input can the pharmacist make to (rejection, calcineurin inhibitor toxicity or ACE maximise the chances of medicine regimen adherence? inhibitor induced). However, in the medium term, diabetic kidney transplant recipients will This patient has a number of potential risk fac- usually receive an ACE inhibitor or angiotensin tors for non-adherence in that she is a single II receptor blocker, even in the absence of trans- person caring for young children, she is blind plant proteinuria. and has a new and potentially complicated Aspirin is usually withheld for a few days medicine regimen about which she currently peri-operatively and reintroduced once the knows very little. 276 Case study answers

The first priority is to simplify JL’s regimen Q9. In addition to co-trimoxazole what other into twice daily tablet dosing, the timing of prophylaxis might you expect to see on this which can be fitted into her life. Medicines can recipient’s TTA? be taken at mealtimes in the same way as she Three months of CMV prophylaxis is likely as manages with her insulin. the donor was CMV positive and the recipient JL needs to be educated about her new med- was known to be CMV negative 3 years earlier, icines. She needs to know the basics of what making the likelihood of primary infection and each medicine is, how she should take it and disease very high. Most transplant centres will to develop a strategy for tablet/capsule recogni- repeat the recipient’s CMV IgG status when tion. This might be best served with a moni- bloods are taken immediately before the trans- tored dosage system (MDS) if it is her wish. Care plant. If this shows that the patient has sero- must be taken to ensure that tacrolimus is converted between transplant listing and being not taken before clinic visits. In addition JL called then the choice of whether to continue should be aware of those medicines such as with prophylaxis is centre-specific. With an the prophylactic antibiotics which have a fixed improving renal function at the point of dis- duration. charge it is imperative that the dose of antiviral She must then be given as much information continues to be correlated with graft function. as she needs to understand the implications of Antifungal prophylaxis might also be consid- the medicines. She can pass written information ered for the early period of transplant immuno- to a family member if desired. If this is satis- suppression. factory for her, and she can participate in treat- ment decisions, her adherence can be as full as is clearly the case with her diabetes care. Chapter 10

Q7. If the transplanted kidney continues to Q1. Is the dose of flucloxacillin reasonable for a function poorly what could be the possible haemodialysis patient? reasons? The SPC for flucloxacillin states that its use in The most likely reasons would be ongoing patients with renal impairment does not usually steroid-resistant rejection, recoverable acute require a dose reduction. However, it also states tubular necrosis (ATN) secondary to either an that in the presence of severe renal failure acute rejection or a nephrotoxic level of (creatinine clearance <10 mL/min) a reduction tacrolimus or lastly a surgical issue such as graft in dose or extension in dosage interval should thrombosis. be considered. As described in the text previ- ously, patients undergoing intermittent haemodialysis should be considered in the cat- Q8. What treatment options would be typical? egory of CrCl <10 mL/min when choosing drug In such a scenario it is most likely that the graft doses. Although it has stipulated a creatinine would be again be re-biopsied to define the clearance for ‘severe’ renal failure, the SPC does problem. Ongoing rejection despite steroid not indicate how to reduce the dose or change would require treatment with rATG or the dosage interval. From the SPC we can also muromonab-CD3. ATN would require ‘watchful see that the usual IV dose for flucloxacillin is waiting’. 250 mg—1 g four times daily, doubled in severe Even in light of a known rejection episode a infection. Following an IV dose, 76.1% is recov- dose adjustment of tacrolimus is necessary for ered in an active form in the urine and a small a level of 18 ng/mL. Dose adjustments should proportion of the dose is excreted in the bile. be proportional to the desired level; if 12 mg/ Excretion of flucloxacillin is slowed in the pres- day results in a level of 18 ng/mL then it is rea- ence of renal failure. sonable to assume that 9 mg/day will result in Evidently the excretion of flucloxacillin will the target range of between 12 and 14 ng/mL be affected by renal failure but as specific dosage Case study answers 277 instructions are not given in the SPC it is pru- needs to be monitored for therapeutic efficacy. dent to consult other available texts. The Renal His temperature and white blood cell count Drug Handbook suggests dosing for haemo- should be regularly monitored as well as the dialysis patients should be the same as those for clinical symptoms. If he does not improve a normal renal function up to a maximum of 4 g change of antibiotic and/or removal of his per day.18 haemodialysis line will be required. As Mr PT has a sepsis and the source is The administration of IV iron to Mr PT dur- thought to be the tunnelled line needed for ing his illness should also be reviewed as IV iron haemodialysis it is important to treat quickly may worsen his infection. and to get high therapeutic levels. The conse- quences of undertreating are a deterioration of Mr PT’s health due to sepsis and the removal of Chapter 11 his haemodialysis line. It would therefore seem appropriate to use the dose of 1 g four times Q1. What are the metabolic abnormalities? daily. The metabolic abnormalities exhibited by the patient are: Q2. Should supplementary doses be given on dialysis days? • Acute renal failure • Hypokalaemia Supplementary doses should only be used in • Hypomagnesaemia haemodialysis if significant drug removal takes • Hypocalcaemia. place during the haemodialysis and where there is a long delay before the next dose is due. The When all electrolytes are reduced like this, it SPC states ‘flucloxacillin is not significantly suggests a renal tubular leak syndrome. removed by dialysis and hence no supple- mentary dosages need to be administered either Q2. What has caused these abnormalities? during or at the end of the dialysis period.’ This The cause of these abnormalities is almost cer- could also have been predicted from the tainly cisplatin. pharmacokinetic data as flucloxacillin is 95% protein bound and therefore significant removal • Cisplatin causes acute renal impairment and is unlikely. a tubular leak syndrome by affecting the S3 segment of the proximal tubule. Thirty per cent of patients are affected. Most recover but Q3. How should Mr PT be monitored? up to 20% can be left with a long-term reduc- Mr PT has been prescribed a high dose of flu- tion in renal function. Biopsies of these cloxacillin for someone receiving haemo- patients show tubular fibrosis. Up to 30% dialysis, therefore he should be monitored for of patients can have long-term magnesium signs of toxicity and adverse effects. The SPC wasting. can be consulted for the whole list of side- • Prednisolone is not nephrotoxic. effects but the most important ones are: CNS • Methotrexate is nephrotoxic, especially in toxicity including convulsions; hepatitis and high doses, where it can precipitate in the cholestatic jaundice (liver function tests should tubules. It can produce a transient reduction therefore be monitored); and diarrhoea and in GFR during infusion but this generally antibiotic-associated colitis. Any of these would recovers immediately on stopping the drug. necessitate a change in therapy. It is also It is almost entirely renal excreted and needs important to note that the flucloxacillin injec- dose reduction in renal failure to reduce tion has a sodium content of 2 mmol/g. This toxicity. will be important in his fluid assessment and • Bleomycin is not nephrotoxic but needs dose interpretation of blood results. reduction when the GFR is <25–35 mL/min As important as measuring toxicity Mr PT (i.e. moderate renal impairment). 278 Case study answers

• In addition, amongst other chemothera- some tumours. Oxaliplatin may be as effec- peutic agents, cyclophosphamide can cause tive but less toxic. haemorrhagic cystitis and lead to bladder • Amifostine is an organic thiophosphate and malignancy in future. Ifosphamide can give can donate a thiol group to reduce toxicity. haemorrhagic cystitis but also a Fanconi-like It has only been tested in high-dose platinum syndrome with metabolic acidosis, hypo- therapy in ovary and small cell lung carcin- kalaemia, hypophosphataemia, nephrogenic oma, but it reduces nephrotoxicity. diabetes insipidus, aminoaciduria and renal impairment. Q4. What has happened now? • Almost all chemotherapy agents can induce a haemolytic uraemic syndrome. Mr NT’s pre-existing renal impairment from his • In cases where the patient has a large tumour previous course of chemotherapy has predis- mass, you can get a tumour lysis syndrome, posed him to damage from further renal insults. where the breakdown products from tumour The subsequent course of cisplatin has now cell destruction can block the filtering sys- caused the patient to develop cisplatin-induced tem in the kidneys, causing acute renal renal impairment. failure. By day 4 he already has severe renal impair- ment (calculated creatinine clearance of 16 mL/min using the Cockcroft and Gault Q3. How can you avoid this complication? equation) and he will require renal replacement therapy should his renal function deteriorate This complication can be avoided in future by: further. • Calculating Mr NT’s body surface area (BSA) using his ideal body weight rather than his Q5. Comment on the antibiotic therapy he has actual body weight. His BSA using his actual been prescribed. Do you need to intervene? weight of 101 kg is 2.09 m2. However, calcu- lating his ideal body weight to be 64 kg, his This is not the best choice of antibiotics for BSA in this instance would be 1.72 m2 . This neutropenic sepsis and it is probably better to would have a marked impact on the dose of liaise with microbiology for a better combina- cisplatin he would be given, and dosing tion. However, in the case of this particular according to his ideal body weight BSA combination, several of them are renally would significantly reduce the risk of excreted, therefore will accumulate in patients nephrotoxicity. with impaired renal function, and cause severe • Fluid loading with sodium chloride. Fluid side-effects. Hence, it will be necessary to mod- loading increases the renal clearance of these ify several of the dosage regimens. Note, if a agents by enhancing urine output, but more drug is renally excreted and accumulates, the importantly a high chloride concentration toxicity seen is not necessarily nephrotoxicity, can reduce the toxicity of the platinum drug. but an exaggeration of whatever the symptoms Cisplatin is toxic in cells with low chloride may be in overdosage of this drug, for example, concentrations by forming reactive hydroxyl neurotoxicity, bone marrow suppression, etc. radicals with water. Therefore increasing chloride will reduce toxicity and some sug- Q6. What would you recommend? gest giving cisplatin in hypertonic saline (e.g. For the existing prescription, the following sodium chloride 1.8%). points should be taken into account: • Choosing an alternative chemotherapeutic agent. Carboplatin is said to be less toxic, • Metronidazole – metabolised by liver, no dos- especially since the dose is based on the area age reduction necessary in renal impairment. under the curve (AUC) exposure to the drug, • Ceftazidime – renally excreted, and toxic in and so automatically adjusts for renal impair- overdose (mainly neurotoxicity). Must ment, but it may also be less effective with substantially reduce the dose in renal Case study answers 279

impairment. Suggest 1 g three times or twice three doses then monthly for six months. This daily in Mr NT and monitor response. is the dose adjusted for GFR <10 mL/min. • Amikacin – exclusively renally excreted, and Cyclophosphamide is renally excreted and extremely nephrotoxic and ototoxic. Under requires dose adjustment. A further reason for no circumstances should the full dose of dose reduction would be the patients age as 7.5 mg/kg twice daily be attempted, as has elderly patients are more susceptible to adverse been prescribed. With Mr NT’s current level events. of renal function, suggest giving a stat dose of 2 mg/kg, waiting 24 hours and doing ran- Q3. How would you monitor this therapy? dom trough level, then dosing again as nec- essary, when the trough is less than 5 mg/L. Cyclophosphamide suppresses the bone marrow This dose will need to be reviewed should Mr so a full blood count including neutrophils is NT be commenced on renal replacement imperative. The nadir for bone marrow suppres- therapy. sion is 10 days after administration so this would be the best time to take a blood sample. Also note that the patient’s renal function is Cyclophosphamide may also affect liver func- deteriorating rapidly, therefore using the tion so liver enzymes should also be checked. Cockroft and Gault equation to estimate renal Prednisolone therapy can cause hyperglycaemia, function will be unreliable. It will be necessary lipid abnormalities and hypertension so these to reassess the patient’s renal function each day, parameters should be checked regularly. A bone and adjust drug dosages accordingly. density scan may be appropriate if the pred- Other possible antibiotics that could be pre- nisolone can not be tapered quickly and long- scribed for neutropenic sepsis include: term therapy is required. Success of treatment is • Meropenem – again, this drug is excreted via measured by a resolution of symptoms, the renal route, so the dose will need to be improvement in renal function and a reduction reduced in severe renal impairment. in c-ANCA titre. • Tazocin – this drug has dual liver and renal excretion, but the dose should be modified Q4. What adjunctive therapies would you slightly in severe renal impairment. consider for this patient? • Oral mesna for prevention of urothelial Chapter 12 toxicity given 2 hours before, 2 hours after and 6 hours after the cyclophosphamide Case 1 infusion. • Metoclopramide should be prescribed for Q1. What diagnosis is most likely to be made? nausea. Ondansetron is a suitable alternative The most likely diagnosis is Wegener’s granulo- if metoclopramide is ineffective. matosis. The presence of c-ANCA and granulo- • Co-trimoxazole for prevention of Pneumo- matous lesions distinguish it from other types of cystis jiroveci (P. carinii) pneumonia. Dapsone vasculitis. Haemoptysis is likely to be due to may be used in patients allergic or intolerant pulmonary haemorrhage which is also more to co-trimoxazole. common in Wegener’s granulomatosis. • Proton pump inhibitor such as omeprazole 20 daily to reduce the risk of gastric ulcera- tion while on concomitant prednisolone. Q2. What initial treatment would you • Osteoprophylaxis with calcium and vitamin recommend? D supplements should be considered. Initial treatment should be IV pulsed methyl- Bisphosphonates such as alendronate 70 mg prednisolone 1 g daily for 3 days. Followed by weekly should also be considered though oral prednisolone 1 mg/kg/day. Then IV cyclo- this is unlicensed in patients with a GFR phosphamide 7.5 mg/kg every two weeks for <10 mL/min. 280 Case study answers

Q5. What advice would you give to the patient dition is deteriorating quite rapidly so further regarding their new treatment regimen? efforts are required to arrest the decline in renal • Patients taking immunosupression are at function as the alternative may be dialysis! An higher risk of infection and should be suitable regimen would be oral cyclophos- advised to seek medical advice at the onset of phamide 2 mg/kg/day together with predniso- any signs of infection such as coughs and lone 1 mg/kg/day. The prednisolone dose may colds. be tapered gradually provided there is no deteri- • Cyclophosphamide may cause thrombo- oration in symptoms. There is no consensus on cytopenia so patients should be asked to the length of course. report any unexplained bruising or bleeding. • Patients should be asked to carry a steroid Chapter 13 card and advised to avoid contact with people with chickenpox. Q1. How would you determine the baby’s GFR? • The importance of complying with immuno- suppressive therapy should be stressed as, in It is very difficult to determine the GFR accur- addition to the risk of disease progression, ately. Normal GFR based on age can be deter- 2 relapses require further immunosuppressive mined by Fawer’s table = 13 mL/min/1.73 m . therapy which carries complications. However, as the baby’s creatinine is measured as 234 µmol/L (normal range 53–97 µmol/L) it can be seen that the baby is in chronic renal failure. Case 2 Q2. Describe the therapy that should be Q1. What drug therapy would you suggest? initiated and why. First-line therapy for any patient with protein- The baby should be started on chronic renal uria is ACE inhibition. A suitable starting regi- failure medicines. As the phosphate is above men would be ramipril 1.25 mg daily increasing 2 mmol/L, calcium carbonate as a phosphate every few days in increments of 1.25–2.5 mg binder should be prescribed. Start with a small according to blood pressure. dose (125 mg) given with each 4-hourly feed and adjust as necessary. Calcium level is in the Q2. What is the target blood pressure for this normal range but the baby has a very high PTH patient? at 10.0 pmol/L. To ensure strong and healthy bones alfacalcidol should be prescribed, but the The target blood pressure in patient with pro- serum calcium needs to be monitored. teinuric renal disease is <125/75 mmHg. The haemoglobin is 12 g/dL. Oral iron and folic acid should be prescribed. The kidneys will Q3. What is the prognosis for Mr Y? be unable to produce erythropoietin so epoetin therapy should be initiated at 100 units/kg SC Mr Y’s renal function has deteriorated despite once a week. The ferritin level will need to be maximal supportive treatment. Hypertension measured. and proteinuria have also persisted which are all Monitoring of U&Es should continue on a poor prognostic markers. Progression of kidney daily basis at this early stage and adjustments to disease is likely to continue if further interven- therapy made as necessary. tions are not made. It may be appropriate to Blood pressure and fluid balance should be start an immunosuppressive regimen. measured daily.

Q4. What further drug therapy would you Q3. How would you ensure that the child recommend? maintains optimum growth and development? Although evidence for immunosuppression in The baby must be referred to a dietician who IgA nephropathy is not definitive, Mr Y’s con- will advise on the best feeds whilst encouraging Case study answers 281 mum to continue to breastfeed. The urea is in before results accurately reflect renal function. If the high range of normal so the dietician must you use the serum creatinine results and the improve nutrition to reduce this. The baby must Cockcroft and Gault formula you will calculate not gain weight in the form of fluid. Regular an estimated creatinine clearance of between 80 measurement of weight (daily), length and head and 85 mL/min, but as previously explained circumference must be made and plotted on a this information is likely to be misleading. This growth chart. is because MF has stopped passing urine, effec- Tube feeding may be required if vomiting tively meaning that he is in ARF, and his creat- becomes a problem. inine clearance is more likely to be less than 10 mL/min. Q4. The parents are told that the baby will In the early stage of disease all dosing deci- need to start dialysis in the very near future, sions should therefore be based on a clinical and will ultimately require a transplant. The assessment, and creatinine clearance calcula- parents are very keen to donate a kidney as tions only used once a steady state has been soon as possible. They are aware of the work up reached. This typically occurs within 3–4 days of required, but what is the earliest age that a admission, depending on the speed of onset and transplant could go ahead and why? the severity of the renal failure. Before a transplant can be considered all vac- The dosage of drugs should be confirmed in cinations must be up to date. An accelerated standard sources. Additionally, in critically ill immunisation schedule can be carried out but patients you need to ensure that therapeutic the earliest age at which a child can be trans- levels of antibiotics are achieved quickly by planted is 19/20 months. using a loading dose. This usually means giving a typical maximum single dose (e.g. cefotaxime 2 g, and 2.4 g benzylpenicillin). Thereafter the Q5. What is the likely diagnosis and what dosage should be adjusted to allow for the renal would you therefore recommend that mum dysfunction (i.e. cefotaxime 1 g four times daily should do? and benzylpenicillin 1.2 g every four hours). The baby almost certainly has peritonitis. The Renal failure should not affect dosage of baby must be admitted to hospital immediately, inotropic agents such as noradrenaline (nor- and as the baby shows signs of systemic illness epinephrine) and other catecholamines as these IV vancomycin and IV ciprofloxacin should be have very short half-lives and are not renally prescribed. A PDF sample should be sent to cleared. These drugs are always given according microbiology and the peritoneal dialysis regi- to response. men should be changed to continuous cycling. Treatment will then be modified according to microbiology results. Q2. The standard protocol for sedating patients on the ICU uses a combination of morphine and midazolam. The clinical staff are Chapter 14 concerned about drug accumulation with these agents. What are the options to prevent Q1. Assess his renal function, and decide accumulation? whether it will be necessary to adjust the Morphine and midazolam are standard sedative dosage of his antibiotics and any other agents in many ICUs but are more likely to medications. cause oversedation in renal failure due to accu- In the acute phase of illness, reviewing clinical mulation of active metabolites. There are vari- findings, particularly urine output, will often ous options to prevent oversedation. One is to give a better guide to renal function than use regular (daily) sedation holds when the laboratory tests. This is because creatinine and sedation is stopped to offset the risk of accu- other markers of renal impairment take time to mulation. Alternatively non-accumulating accumulate. Consequently there is a lag time agents such as fentanyl or alfentanil for 282 Case study answers analgesia, and propofol or lorazepam for anaes- additional anticoagulants as this will increase thesia could be used. These medicines tend to the potential for haemorrhagic side-effects. be more expensive but this additional cost Drotrecogin alfa will also provide more than maybe cancelled out as delayed recovery from adequate anticoagulation for CVVHF. sedation is less likely. The downside is that clin- ical staff maybe unfamiliar with these agents Q5. What options are available to manage MF’s and therefore use them inefficiently, resulting in metabolic acidosis? poor-quality sedation if too little is given, or MF’s metabolic acidosis is probably due to a accumulation if too much is given. In practice combination of renal impairment and anaerobic there is no right or wrong drug choice, the key metabolism secondary to organ hypoperfusion issue is to use the chosen agents carefully. and sepsis. This anaerobic metabolism is reflected by the raised blood lactate level which Q3. The CVVHF system is configured to an is due to excess lactate production and an ultrafiltration rate of 30 mL/kg/h. Are any inability to metabolise this to bicarbonate. The dosage modifications required for the drugs best method to correct this acidosis is to offset mentioned above and for the following the causes (e.g. improve tissue perfusion by use additional agents – enoxaparin SC 40 mg once daily, ranitidine IV 50 mg three times daily, of inotropes and fluid resuscitation) and the and IV insulin infusion to maintain a blood use of CVVHF. In this case a bicarbonate-based glucose of 5–8 mmol/L? haemofiltration fluid should be chosen as the replacement fluid as opposed to a lactate-based An ultrafiltration rate of 30 mL/h/kg is equiva- fluid. The latter will probably aggravate the lent to 30 70 (i.e. 2100 mL/h or 35 mL/min). already raised lactate levels. This is maximum drug clearance that can be achieved by the CVVHF device. As for all other dosage decisions you should always refer to Chapter 15 standard sources, but with drugs that are renally cleared you will also need to consider the Q1. Comment on this prescription. What CVVHF clearance. In this case the following interventions might you make? dosages are suitable for a patient with ARF Prescription for amitriptyline and tramadol MR. receiving 35 mL/h CVVHF clearance. Things to consider: • Cefotaxime 2 g four times daily • Tramadol, amitriptyline and fluoxetine can • Benzylpenicillin 1.8 g every 4 hours all increase serotonin levels, therefore using • Sedation, insulin and inotropes – according all three together gives much increased risk to response of developing serotonin syndrome. Therefore • Ranitidine 50 mg twice daily this combination is best avoided. • Enoxaparin 40 mg once daily. • Tramadol, amitriptyline and fluoxetine can Note: Enoxaparin thromboprophylaxis is usu- all lower the seizure threshold, therefore ally omitted as the patient will probably be using all three together increases the risk of receiving anticoagulation to facilitate the this occurring CVVHF. Typical agents used are unfractionated • Tramadol is 90% excreted in urine and little heparin or epoprostenol. is removed by dialysis. Use a non-MR (modi- fied release) preparation at 50 mg twice daily in ESRF. Q4. Are any dosage modifications required for • Tramadol is converted to its active form (M1 this agent, and are any additional precautions metabolite) in the body. It has been proposed necessary? that SSRIs block this conversion process, Drotrecogin alfa (activated protein C) is cleared therefore rendering tramadol inactive. This systemically so no dosage adjustment is neces- would be another reason for not using these sary. A more important concern is to stop any two drugs together. Case study answers 283

• In some cases an SSRI and a TCA can be used Kumamoto studies to reduce the risk of devel- together. This would usually be in a situation oping microvascular complications. where a patient had neuropathic pain that was responding to a TCA and was also depressed. The first option would be to try Q4. What might her causes of acute renal failure be? and increase the TCA to an antidepressant dose, but often this is not tolerated. Then the Sepsis, drug-induced (metformin and or val- next step would be to try an SSRI. If the two sartan) or hypovolaemia. classes of drugs were being used together then the dose of amitriptyline would be limited to a maximum of 75 mg once daily. Q5. Explain her drug history and any possible reasons for stopping some of her medication on (SSRIs are thought to inhibit the metabolism admission. of TCA by cytochrome P450 CYP2D6 result- ing in increased serum concentrations of Triple OHA therapy, as mentioned in the chap- the TCA.) ter, may be beneficial for a 2–3 month period as glitazones can take several months to show their full effect, although initiating insulin Suggestions for treatment: treatment should not be delayed if there is • Consider using gabapentin at reduced dosage pancreatic beta cell exhaustion. Metformin, as instead of amitriptyline. discussed earlier, is safe to use in chronic • Consider an alternative analgesic to tramadol renal failure as long as the GFR is above (e.g. co-codamol 30/500). Would need to dis- 60 mL/min/1.73 m2. In this particular case, it is cuss what the patient had tried before. sensible to stop metformin and withhold rosigli- tazone and glicazide on admission until invest- An important point to note is that the patient igations are done and DP’s blood glucose levels feels no one believes she has pain, as no physical return to above 5 mmol/L and she is able to eat. cause for it has been found. This is quite a com- ACE inhibitors and angiotension II receptor mon scenario; the majority of chronic pain antagonists have been shown to reduce the rate patients do not have a physical cause for their of progression in both diabetic nephropathy pain. An important part of the treatment process and non-diabetic nephropathy, so it seems sen- is to reassure patients that you do believe them sible for DP to be on valsartan. What is not and try to help them understand the reasons why known is whether this has caused the acute their pain is occurring. renal episode, so bilateral renal artery stenosis needs to be ruled out, but also DP has presented with extremely low BP. ACE inhibitors/ Chapter 16 angiotension II receptor antagonists can also cause hyperkalaemia and as DP presented with Q1. What might her causes of diabetes be? high potassium the valsartan is stopped. Hypertension and/or obesity. Beta-blockers should be used with caution in diabetes as they may mask hypoglycaemia, but it is not a contraindication to treatment. NICE have Q2. What might her causes of chronic renal recently published an update for hypertensive failure be? treatment and no longer sanction their use unless Poorly controlled blood glucose and blood no other alternative can be found. DP is a long pressure. standing patient so it is not unreasonable for this patient to present on this treatment, although it may be an opportunity to trial an alternative Q3. Could it have been prevented? agent when her BP begins to normalise. Yes, tight blood glucose and blood pressure has Flucloxacillin and cefalexin initiated by the been proven by the DCCT, UKPDS and GP for cellulitis are known to cause interstitial 284 Case study answers nephritis, so this may be another cause of acute BP control to ≤135/75 mmHg. Beta-blockers, renal failure, although the likelihood is low. also mentioned before, are not contraindicated Cephalosporins can also cause diarrhoea, so this in diabetes but this may be an ideal opportunity may be the culprit for DP’s presenting diarrhoea. to recommend an alternate agent. The patient’s BP on discharge was below the recommended target so at this stage another antihypertensive Q6. Why would this patient take atorvastatin agent is not necessary, but a recommendation of and aspirin? a calcium channel blocker could be noted for The major causes of death in type 2 diabetes are the follow-up plans. cardiovascular-related illnesses, so all patients with diabetic kidney disease should take a low- Q10. What discharge plans should be made for dose aspirin and a lipid-lowering agent for vas- this patient? cular protection. The patient has been switched from OHA ther- apy to injecting insulin therapy, which is a huge Q7. Why change this patient’s diabetic regimen change for DP, so she will need education and to insulin? training for insulin administration and monitor- DP has a history of poorly controlled blood ing from a diabetic link nurse. Education on diet glucose levels and a HbA1c result of 9% despite and exercise will also be necessary due to the triple OHA therapy. This may be a result of pan- increased side-effect of weight gain with insulin creatic beta cell exhaustion. If there is some therapy. Awareness of hypoglycaemia needs residual insulin secretion then an option for DP emphasising to DP again due to the increased could be a daytime metformin with a nocturnal incidence with insulin therapy. Follow-up within insulin, although this is probably not an option the community for blood glucose and blood due to the failure of the triple OHA therapy. The pressure monitoring will also need to be organ- best treatment at this stage for DP is a twice a ised as well as regular screening for ESRF. day insulin regimen, as recommended by the diabetologist with careful monitoring for hypo- glycaemia and weight gain. Chapter 17 Q1. He has been transferred to your ward. As Q8. Would the regimen differ if the patient had the ward pharmacist, what further information ESRF? would you like to know? In ESRF the risk of hypoglycaemia is higher as Drug history – pre-admission, during hospital the kidney, with the liver, metabolise circulating stay, any allergies. insulin, so in kidney impairment or failure this will result in an accumulation of this circulating Q2. At the previous hospital he was given insulin and thus hypoglycaemia. In ESRF the gentamicin, vancomycin and pamidronate? insulin regimen may require a dose reduction or Could any of these contribute to his renal a switch to a once a day basal human analogue failure? such as glargine or levemir. Gentamicin, vancomycin and pamidronate can all cause ARF. Q9. Comment on BP control in diabetes. Q3: What would be your pharmaceutical care As already discussed, ACE inhibitors and plan at this stage? angiotensin II receptor antagonists are benefi- cial in diabetic nephropathy and as there is no See ARF pharmaceutical care – avoid nephro- renal vascular disease restarting DP on her val- toxic drugs, check fluid status, and consider the sartan is sensible but at a lower dose with a rec- need for further therapy of hypercalcaemia and ommendation to titrate the dose to maximise the need for antibiotics. Case study answers 285

Q4. Would your pharmaceutical care plan Q4. What would you discuss at this stage? What change at this stage? Outline a new medications could you ensure were prescribed pharmaceutical care plan. for the last few days of life? • Ensure that the correct doses have been pre- Discuss conservative management, including scribed – any requirement for modification in active symptom management, analgesia, view of renal impairment? antiemetic, sedative and glycopyrronium. • Ensure appropriate supportive therapy is prescribed • Timings of doses relative to dialysis Chapter 19 • Monitor for adverse effects • Patient counselling. Q1. What advice would you give him? For each patient the renal diet is different. It Q5. After 10 days Mr CB remains depends on the patient’s normal dietary intake, haemodialysis-dependent as his renal function stage of kidney disease, biochemistry and treat- has not improved. The renal team would like to ment. Dietary restriction may include protein, treat Mr CB’s anaemia and commence an phosphate, potassium, sodium and fluid. As his erythropoesis-stimulating agent. What dose biochemistry is within acceptable ranges and his would you recommend? Are there any other weight is fairly steady, only healthy eating, low factors to consider? salt advice would be given. If Mr Smith is over- Patients with anaemia due to bone marrow infil- weight then advice to reduce his weight, lead- tration, chemotherapy and kidney disease may ing to a decrease in blood pressure, and some require higher doses of chemotherapy. Any of lifestyle advice should be given. This would the currently licensed erythropoesis-stimulating include advice about increasing exercise and agents may be given – see individual summary decreasing the amount of salt within his diet by of product characteristics for detailed dosing avoiding convenience foods and by cooking advice. with fresh ingredients. If ready-made foods are the only way to ensure that a patient’s nutri- tional status is not compromised then the Chapter 18 patient should be advised to look at the food labelling and choose products that contain Q1. What are the possible causes of his pain? 0.25 g salt (0.1 g sodium) or less per 100 g. His biochemistry, weight and appetite should The pain in the left leg could be due to diabetic be monitored closely. neuropathy or metastases, and the pain in his shoulder is likely to be due to metastases. Q2. What advice would you give him now? Q2. How would you manage his nausea and Advice would be give to limit the dairy foods – vomiting? milk, cheese, yoghurts, offal meats, nuts, fish Adding in metoclopramide will act as an with edible bones and shellfish – ensuring that antiemetic and also help diabetic gastroparesis. he was meeting his protein and energy require- Dose reduction in end stage renal disease is rec- ments to prevent malnutrition. ommended, although normal doses are used regularly in practice. Q3. What further dietary measures would be needed? Q3. What might be causing his pain? What First check for non-dietary causes of hyper- other treatment would you consider to relieve kalaemia, such as drugs or blood transfusions. his pain? Second, review the diet to check for non- The pain is likely to be due to restless legs (see nutritious potassium foods (use of salt substi- Table 18.6). tutes, excessive intake of fruit and fruit juice, 286 Case study answers vegetables and potatoes that have not been traveller such as Ellen. This should be discussed boiled in water). It is essential at this stage to with travel health experts and the supervising ensure the diet remains nutritionally adequate clinician. If she travels unprotected by the vac- so a full dietetic assessment is necessary. cine, she should minimise her risk of yellow fever transmission by remaining on the ship in Q4. Which supplements would you suggest? Colombia and taking good insect bite precau- tions during daylight hours if on deck/land. A Supplements chosen would depend on the yellow fever certificate would be required for patient’s nutritional requirements, biochem- onward travel to Barbados (who require certifi- istry, other medical history – diabetes, patient cate of vaccination from all travellers arriving likes and dislikes. The basic 1.5 kcal/mL milk- from, or passing through, endemic areas). If based supplements are Ensure Plus, Fortisip, Ellen is unable to receive the vaccine, she would Fresubin Energy and Resource shake. Other require a letter of medical exemption from the options include fruit juice-based supplements – vaccination. This may be taken into considera- Enlive, Fortijuice (which are higher in glucose tion by port officials but should not be consid- than the milk-based supplements). Supplements ered an automatic waiver. not recommended for renal patients are Build- Ellen’s response to any vaccination may be up and Complan. Food fortification is also rec- suboptimal. Food and water hygiene and insect ommended with fats (fried foods, olive oil, bite precautions should be emphasised. double cream, Calogen) or with sugar (fizzy drinks, glucose-based polymers – Vitajoule, Q3. What malaria prophylaxis is appropriate? Maxijul). Always ensure that the patient is referred to Ellen is travelling to low-risk areas for malaria a dietician so a detailed nutritional assessment and so does not require chemoprophylaxis. Full can be carried out and the patient is monitored. bite protection measures, however, should be taken. The importance of recognising signs and symptoms of malaria should also be discussed Chapter 20 with Ellen. She should be advised to seek rapid medical attention if she develops any flu-like Case 1 symptoms, fever, or any other unexplained symptoms, 7 days or more after entering a Q1. What are the vaccinations required for malarial area and for up to one year after exit. travel to these areas?

Check that Ellen’s British schedule is up to date. Case 2 A tetanus booster may be required if her last dose was more than 10 years ago. The Americas Q1. What would be appropriate malaria are considered polio free, so polio vaccination is prophylaxis for travel to these areas? not required provided Ellen is fully vaccinated according to the British schedule. Pneumocco- Dorothy is travelling to Kruger (South Africa), cal, influenza, hepatitis A and yellow fever (for Zambia, and Botswana which are risk areas for Colombia) are normally advised where not med- falciparum malaria. The Advisory Committee on ically contraindicated (see below: caution with Malaria Prevention recommends mefloquine, live vaccines). doxycycline, or Malarone.

Q2. What other considerations need to be taken Q2. Which would be the most appropriate into account when recommending appropriate chemoprophylaxis for Dorothy? vaccinations for Ellen? Mefloquine would be the most suitable option. Yellow fever is a live vaccine which would not Malarone (atovaqone/proguanil) is contra- be recommended to an immunocompromised indicated for severe renal failure. Dorothy’s Case study answers 287 calcium-based phosphate binder may interact Zambia was declared yellow fever free by the with doxycycline and reduce its antimalarial World Health Organization in 2002 and effects. Dorothy is not at personal risk of this disease for this itinerary.

Q3. What advice about malaria prophylaxis should be given to Dorothy? Case 3

Dorothy should be advised to ideally start tak- Q1. What would be appropriate malarial ing the mefloquine three weeks before travel. prophylaxis for travel to Punta Cana? She should take it at the same time each week, for the duration of her trip and for four weeks There is a risk of malaria in the Dominican after leaving Africa. The importance of taking Republic, including recently (2005) to tourist the medication regularly and for the full dura- travellers who have only stayed in coastal tion should be emphasised. She should also be areas. Chloroquine or proguanil are usually informed about the possible adverse effects of recommended. mefloquine, and to seek medical advice should they occur so a suitable alternative can be Q2. Which agent would be most appropriate for prescribed. Mary? It is important that Dorothy understands the Neither drug is ideal for Mary. Proguanil should need for scrupulous mosquito bite precautions be avoided due to her previous reaction to the in addition to chemoprophylaxis. drug. If chloroquine is prescribed there is an Dorothy should also be made aware of the increased risk of ciclosporin toxicity, as the signs and symptoms of malaria. She should plasma concentration of ciclosporin can be be advised to seek rapid medical attention if she increased by chloroquine. Careful monitoring of develops any flu-like symptoms, fever, or any ciclosporin levels would be required and dose other unexplained symptoms, 7 days or more adjustments made accordingly. after entering a malarial area and for up to one year after exit. Q3. Would any other antimalarial be suitable?

Q4. Which vaccinations are required for travel Other agents used for malaria chemoprophyl- to these areas? axis are Malarone, mefloquine and doxycyline. Malarone contains proguanil so is not an Check Dorothy’s British schedule is up to date option. Mefloquine is contraindicated due to and give booster doses of tetanus and polio if Mary’s previous medical history of post-natal her last doses were more than 10 years ago. depression. Pneumococcal, influenza, hepatitis A, hepatitis Doxycycline, like chloroquine, may interact B and typhoid are normally recommended. Her with ciclosporin and increase the plasma con- holiday dialysis unit will also usually require centration of ciclosporin. Close monitoring Dorothy’s hepatitis B immune status. Rabies with appropriate ciclosporin dose adjustments vaccine could also be offered, although it is not would be required to prevent toxicity. generally suggested for short trips. Dorothy should be given advice about risk avoidance and Q4. What advice would you give to Mary? the importance of post-exposure treatment for rabies, which is required whether or not pre- Mary should consider the option of changing exposure vaccine has been received. her holiday to a non-malarious destination.

Glossary

Absolute iron deficiency Marked by low necrosis, which is a form of cell death that serum ferritin. results from acute cellular injury, apoptosis is Accelerated phase hypertension Essential carried out in an ordered process that gener- hypertension characterised by acute onset, ally confers advantages during an organism’s severe symptoms, rapidly progressive course, life cycle. and poor prognosis. Atherosclerosis An arteriosclerosis charac- Acute tubular necrosis (ATN) The term used terised by atheromatous deposits in and to describe the pathology where tubular cells fibrosis of the inner layer of the arteries have died and have not yet been replaced by Anuria Passing less than 50 mL urine per day. new ones. Adaptive immune system The cellular aspect β -Microglobulin A component of MHC class of the immune systems of higher life forms 2 I molecules, which are present on almost which allow recognition and memory of all cells of the body (red blood cells are a pathogens. notable exception). In patients on long-term Adynamic bone disease Low turnover bone haemodialysis, they can aggregate into disease, where bone cell activity is reduced or amyloid fibres that deposit in joint spaces, absent. In contrast to osteomalacia, there is a disease known as dialysis-related amy- no increase in bones lacking minerals. People loidosis. with adynamic bone disease have a tendency Biocompatible Not having toxic or injurious to develop more fractures and blood vessel effects on biological function. calcification. Allograft A transplanted organ sourced from a non-identical member of the same species. Cachectic A profound and marked state of Amyloidosis Amyloids are insoluble fibrous constitutional disorder; general ill-health and protein aggregations sharing specific struc- malnutrition tural traits. Amyloidosis is defined as any Calciphylaxis An adaptive response that fol- extracellular, proteinaceous deposit exhibit- lows systemic sensitisation by a calcifying ing cross-beta structure. factor (as a vitamin D) and a challenge Anaphylactoid Resembling anaphylaxis or (as with a metallic salt) and that involves anaphylactic shock, i.e. hypersensitivity (as local inflammation and sclerosis with cal- to foreign proteins or drugs) resulting from cium deposition, leading to necrotic skin sensitisation following prior contact with the lesions. causative agent. Cardiogenic shock Shock resulting from fail- Antinociception Reduction in the perception ure of the heart to pump an adequate of pain (nociception). amount of blood as a result of heart disease Apoptosis One of the main types of pro- and especially heart attack. grammed cell death. As such, it is a deliber- Carpal tunnel syndrome Swelling within the ate process of life relinquishment by a cell in carpal tunnel causing pain and numbness in a multicellular organism. In contrast to one half of the hand.

289 290 Glossary

Chronic allograft nephropathy (CAN) functioning of the heart and to diagnose Alternatively termed chronic rejection. The abnormalities and disease. insidious and slow deterioration of trans- Echogenicity Reflecting ultrasound waves. planted kidney function. Ectopic calcification Extraskeletal calcifica- Complex regional pain syndrome A chronic tion, i.e. calcium phosphate deposits in soft progressive disease characterised by severe tissues and blood vessels. pain, swelling and changes in the skin. En-bloc transplant Where both kidneys from C-Reactive protein (CRP) A protein pro- a single donor are transplanted into a single duced by the liver that is normally present recipient. in trace amounts in the blood serum but is Endothelin Any of several polypeptides elevated during episodes of acute inflamma- consisting of 21-amino-acid residues that tion (as those associated with neoplastic dis- are produced in various cells and tissues, ease, chronic infection, or coronary artery which play a role in regulating vasomotor disease). activity, cell proliferation, and the pro- Cryoglobulins Circulating proteins (e.g. IgM) duction of hormones, and that have been which become insoluble at reduced tempera- implicated in the development of vascular tures – less than 4°C. The reaction is revers- disease. ible; redissolution occurs at 37°C. Epoetin Erythropoiesis-stimulating agent Cryoglobulinaemia The presence in the made by various companies that is very sim- blood of cryoglobulin, which is precipitated ilar to natural erythropoietin. in the microvasculature on exposure to Erythrocyte Red blood cell. cold. Erythropoietin Hormone that stimulates Cytolytic enzymes Enzymes which cause the erythropoiesis. death of a cell by bursting, often by osmotic Erythropoiesis The production of red blood mechanisms that compromise the integrity cells (as from the bone marrow). of the cellular membrane. Erythropoiesis stimulating agent ESA – sub- stance that stimulates erythropoiesis (e.g. Delayed graft function (DGF) The temporary epoetin or darbepoetin) and usually recoverable period where a Euvolaemia Normal circulatory or blood fluid patient may continue to require dialysis after volume within the body. a transplant. Extracorporeal circuit Blood circulation in a Darbepoetin An erythropoiesis-stimulating circuit occurring or based outside the living agent made by Amgen. body (e.g. blood passing around a haemo- Dyslipidaemia A condition marked by abnor- dialysis circuit). mal concentrations of lipids or lipoproteins in the blood. Fanconi syndrome A disorder in which the Dysplastic kidneys A condition of both kid- proximal tubular function of the kidney is neys where the tissue is partly normal with impaired, resulting in decreased reabsorption some normal glomeruli but laced throughout of electrolytes and nutrients back into the the kidney is fibrosis (like scar tissue), ab- bloodstream. Compounds involved include normal cell groups such as little pieces of glucose, amino acids, uric acid, phosphate cartilage, and immature tissue where the kid- and bicarbonate. The reduced reabsorption of ney did not finish developing. bicarbonate results in type 2 or proximal renal tubular acidosis, EBPG European Best Practice Guidelines for Fibromyalgia A chronic syndrome charac- renal anaemia. terised by diffuse or specific muscle, joint, or ElectroCardioGram ECG – a trace of the elec- bone pain, fatigue, and a wide range of other trical activity of the heart. symptoms. Echocardiography The use of ultrasound to Functional iron deficiency Marked by trans- examine and measure the structure and ferrin saturation or % hypochromic cells. Glossary 291

Gastroparesis Also called delayed gastric average blood glucose levels over the preced- emptying, a disorder in which the stomach ing 90 days (approximately). takes too long to empty its contents. Is Hepatorenal syndrome Functional kidney usually caused by damage to the vagus nerve, failure associated with cirrhosis of the liver especially in diabetic autoneuropathy. and characterised typically by jaundice, Glomerular filtration The forcing, under ascites, hypoalbuminaemia, hypoprothrom- high pressure, of small molecules such as binaemia and encephalopathy water, glucose, amino acids, sodium chloride Homeostasis The maintenance of relatively and urea, from the blood in the renal affer- stable internal physiological conditions (as ent arteriole across the glomerular basement body temperature or the pH of blood) in membrane of the Bowman’s capsule and into higher animals under fluctuating environ- the nephron. mental conditions. Glomerulonephritis Nephritis marked by Hyperacute rejection The almost immediate inflammation of the capillaries of the renal response to donor antigen where a recipient glomeruli. has preformed antibody. Glycosuria The presence in the urine of Hypernatraemia The presence of an abnor- abnormal amounts of sugar. mally high concentration of sodium in the Granulomatous lesions Small nodules that blood. are seen in a variety of diseases such as Hyperplasia Abnormal increase in the num- Crohn’s disease, tuberculosis and sarcoidosis, ber of normal cells in normal arrangement They are composed of a group of epithelioid in an organ or tissue, which increases its macrophages surrounded by a lymphocyte volume. cuff. Hyperviscosity syndrome An increase in the viscosity of the blood. This may be caused Haematocrit The percentage of the volume of by an increase in serum proteins and may whole blood that is composed of red blood be associated with bleeding from mucous cells as determined by separation of red membranes, retinopathy and especially blood cells from the plasma usually by monoclonal gammopathies such as in mul- centrifugation. A haematocrit ranging from tiple myeloma. An increased viscosity sec- 42% to 52% in males and 35% to 47% in ondary to polycythaemia may be associated females is typically considered normal – also with organ congestion and decreased capil- called packed cell volume. lary perfusion. Haematuria The presence of blood or blood Hypoalbuminaemia Hypoproteinaemia cells in the urine. marked by reduction in serum albumin. Haemodynamic Relating to or functioning in Hypochromic anaemia An anaemia marked the mechanics of blood circulation. by deficient haemoglobin and usually micro- Haemolytic uraemic syndrome (HUS) An cytic red blood cells and associated with lack inflammatory reaction leading to acute renal of available iron. failure (ARF) and disseminated intravascular Hypovolaemia Decrease in the volume of the coagulation (DIC). The fibrin mesh destroys circulating blood. red blood cells and captures thrombocytes, leading to a decrease of both on full blood Immunomodulation Modification of the count. Can be caused by Escherichia coli immune response or the functioning of toxin, HIV, lupus, post-partem, malignant the immune system by the action of an hypertension, scleroderma and cancer immunomodulator. chemotherapy. There is also familial HUS, an Induction immunosuppression Drugs used inherited condition. peri-operatively and in the first few weeks HbA1c Glycosylated haemoglobin – it is prim- after transplant to augment the long-term arily a treatment-tracking test reflecting immunosuppression strategy. 292 Glossary

Intimal proliferation Thickening of the walls Nephrosclerosis Hardening of the kidney; forming blood vessels or renal tubules. specifically: a condition that is characterised by sclerosis of the renal arterioles with Left ventricular hypertrophy (LVH) The reduced blood flow and contraction of the thickening of the myocardium (muscle) of kidney that is associated usually with hyper- the left ventricle of the heart. Disease tension and that terminates in renal failure processes that can cause LVH include any dis- and uraemia. ease that increases the afterload that the Neuropathic pain Chronic pain resulting heart has to contract against (e.g. aortic from injury to the central or peripheral nerv- stenosis, aortic insufficiency and hyperten- ous system. sion), and some primary diseases of the muscle of the heart. Oliguria Passing less then 400 mL urine per Luminal obliteration Shrinkage, leading to day. disappearance of the lumen of blood vessels Osteodystrophy Defective ossification of or tubules within the kidney. bone usually associated with disturbed cal- cium and phosphorus metabolism. Maintenance immunosuppression Oral Osteomalacia A disease of adults that is char- drugs, with different and complimentary acterised by softening of the bones and is mechanisms of action, usually taken in com- analogous to rickets in the young. bination for the life of the transplanted organ. Microalbuminuria The measurement of small Panel reactive antibodies (PRA) A measure amounts of albumin in the urine that cannot of a patient’s level of sensitisation to a stan- be detected by urine dipstick methods, typic- dard panel of donor antigens. ally the excretion of 30–300 mg of albu- Periarticular Relating to, occurring in, or min/24 hours. being the tissues surrounding a joint. Microangiopathic Conditions associated Pericarditis Inflammation of the pericardium, with a disease of very fine blood vessels. the conical sac of serous membrane that Myositis Inflammation of a muscle, especially encloses the heart and the roots of the great a voluntary muscle, characterised by pain, blood vessels. tenderness, and sometimes spasm in the Peritubular cells Cells found adjacent to or affected area. surrounding a renal tubule. Plasma oncotic pressure In blood plasma, Nephropathy An abnormal state of the the dissolved compounds have an osmotic kidney; especially one associated with or pressure. A small portion of the total os- secondary to some other pathological motic pressure is due to the presence of process. large protein molecules; this is known as Nephrotoxic Poisonous or damaging to the the colloidal osmotic pressure, or oncotic kidney, as in nephrotoxic drugs. pressure. NMDA (N-methyl-D-aspartic acid) An amino Plasma exchange A procedure used to separ- acid derivative acting as a specific agonist at ate the plasma from the blood. After plasma the NMDA receptor, mimicking the action of separation, the blood cells are returned to the neurotransmitter glutamate, and associ- the person undergoing treatment, while the ated with learning and memory. plasma, which contains the unwanted anti- Nocturia Urination at night especially when bodies, is discarded and the patient receives excessive. replacement donor plasma or saline with Non-heart beating donor (NHBD) Where the added proteins in its place. organs originate from a cadaver who does Plasmapheresis A procedure used to separate not fall into the typical heart-beating, brain- the plasma from the blood. After plasma stem dead donor category. separation, the blood cells are returned to Glossary 293

the person undergoing treatment, while the Renal artery stenosis Narrowing of the major plasma, which contains the unwanted anti- artery that supplies blood to the kidney. bodies, is first treated and then returned to Renal artery stenosis can lead to seriously ele- the patient. vated blood pressure. Common causes of Pleural effusion An exudation of fluid from renal artery stenosis include atherosclerosis the blood or lymph into a pleural cavity. and thickening of the muscular wall (fibro- Polycystic kidney disease Either of two muscular dysplasia) of the renal artery. hereditary diseases characterised by gradually Renoprotective Protection against damage to enlarging bilateral cysts of the kidney which the kidney. lead to reduced renal functioning. (a) A Residual urine output The amount of urine disease that is inherited as an autosomal still passed by patients. dominant trait, is usually asymptomatic until Reticulocytes Erythrocyte precursors. middle age, and is marked by side or back Reticulo-endothelial system A diffuse system pain, haematuria, urinary tract infections, of cells of varying lineage that include espe- and nephrolithiasis. (b) A disease that is cially the macrophages and the phagocytic inherited as an autosomal recessive trait, usu- endothelial cells lining blood sinuses and ally affects infants or children, and results in that were originally grouped together renal failure. because of their supposed phagocytic proper- Polyuria Renal disorder characterised by the ties. production of large volumes of pale dilute Retroperitoneal fibrosis Proliferation of urine (usually >5 L/24 hours); often associ- fibrous tissue behind the peritoneum often ated with diabetes. leading to blockage of the ureters – also Post-herpetic neuralgia A condition resulting called Ormond’s disease. after nerve fibres are damaged during a case Rhabdomyolysis The destruction or degener- of herpes zoster, causing chronic pain that ation of skeletal muscle tissue (as from trau- may persist or recur for months or years in matic injury, excessive exertion, or stroke) the area affected. that is accompanied by the release of muscle Postural hypotension Symptoms include cell contents (as myoglobin and potassium) dizziness, lightheadedness, headache, blurred into the bloodstream, resulting in hypo- vision and fainting, generally occurring after volaemia, hyperkalaemia, and sometimes sudden standing. It can be caused by blood acute renal failure. pooling in the lower extremities; venous return and cardiac output are further com- Scleroderma A usually slowly progressive dis- promised, resulting in further lowering of ease marked by the deposition of fibrous con- arterial pressure. nective tissue in the skin and often in Pure red cell aplasia (PRCA) A type of internal organs and structures, by hand and anaemia affecting the precursors to red blood foot pain upon exposure to cold, and by cells but not to white blood cells. In PRCA, tightening and thickening of the skin. the bone marrow ceases to produce red blood Serum ferritin Protein that reflects iron cells. Pure red cell aplasia is regarded as an stored as ferritin in tissues. autoimmune disease. Systemic inflammatory response syndrome Purpura The appearance of red or purple (SIRS) An inflammatory state of the whole discolorations on the skin, caused by bleed- body (the ‘system’) without a proven source ing underneath the skin. Small spots are of infection. SIRS can be considered to be a called petechiae, while large spots are called subset of cytokine storm, a general term for ecchymoses. cytokine dysregulation. Pyelonephritis Inflammation of both the parenchyma of a kidney and the lining of Thromboxanes Any of several substances that its renal pelvis especially due to bacterial are produced especially by platelets, are infection. formed from endoperoxides, cause constric- 294 Glossary

tion of vascular and bronchial smooth mus- Vasculitis Inflammation of a blood or lymph cle, and promote blood clotting. vessel. Transferrin Iron carrier protein, reflects avail- Vasculitides A group of conditions charac- able iron. terised by inflammation of blood or lymph vessels. UF coefficient A measure of how permeable the dialyser is. Index

Page numbers in italic refer to figures, tables and boxes.

ABO blood groups, 108–9 anaesthetic agents, sedative, 186 antineutrophil cytoplasmic absorption, drug, 41, 127–8 analgesia autoantibodies (ANCA), 15, acarbose, 209, 211 critically ill patients, 184–7 148, 150 acid/base balance, 5–6 myeloma, 220 antineutrophil cytoplasmic acute interstitial nephritis (AIN), palliative care, 229–33, 231 autoantibody (ANCA)- 141–2, 142 analgesic ladder, 191, 229–33, 231 associated vasculitis, 145, acute renal failure (ARF), 21–34 analgesic nephropathy, 26, 142 148–53 causes, 22–7, 23, 24–5 analgesics, 191–201, 192 anti-thymocyte globulin, rabbit critical care patients, 181–8 angiotensin II, 26, 27 (rATG), 110, 115 dietary management, 31, angiotensin II receptor blockers, anuria, 21, 29, 139 245–7 80, 81, 140, 158, 207 aspirin, 41, 83, 118, 156, 159, 196 drug-induced, 26–7, 139–44 angiotensin-converting enzyme aspirin-exacerbated respiratory myeloma, 217, 218, 222 (ACE) inhibitors, 79–81, 80, disease (AERD), 194 prevention, 181–2 158 atenolol, 166 renal replacement therapy, adverse effects, 26, 89, 140 atovaquone/proguanil (Malarone), 30–1, 182–4 diabetes mellitus, 207 260, 260, 261 acute tubular necrosis (ATN), 23, paediatric kidney disease, atracurium, 186, 187 24, 25, 29 165, 166 atrial natriuretic peptide (ANP), 6 drug-induced, 141, 142 anorexia, 226 autoimmune kidney disease, albumin, serum, 10, 13, 128, 147 anthropometry, 248, 248 145–62, 146 albumin:creatinine ratio (ACR), antibiotics, 92, 97, 100–2, 153 autoregulation, renal blood flow, 205, 207 paediatric patients, 174, 175, 3–4, 22, 139, 139 alcohol consumption, 79 176, 177 azathioprine aldosterone, 6, 11, 13 travellers, 255, 256 autoimmune disease, 150, alemtuzumab, 111 anticoagulants, 142 152, 156 alfacalcidol, 66, 166–7 renal replacement therapy, renal transplant recipients, alfentanil, 186, 187, 197, 230–2, 90, 94, 184 107, 112–13, 117 231 renal transplant recipients, allopurinol, 116–17, 120, 143, 152 120 basiliximab, 110–11 aluminium hydroxide, 64, 89–90 anticonvulsant drugs, 120, 200–1, BCG vaccine, 169, 170, 171 aluminium toxicity, 89–90 201 belatacept, 115 amikacin, 176 antidepressants, 199, 199–200 Bence Jones proteins, 16, 217 aminoglycosides, 101, 102, 117 antidiuretic hormone (ADH), 5, 6 benzodiazepines, 186, 187, 228, amitriptyline, 192, 199, 200 antiemetics, 40, 228–9, 229 234 β amlodipine, 166 antifungal agents, 102, 153, 177 2-microglobulin, 86, 89, 95, 96 amphotericin, 102, 117, 177 antihistamines, 40, 234 beta-blockers, 80, 80, 81, 82, 120 amyloidosis, 89, 219 antihypertensive drugs, 38, 79–81, paediatric kidney disease, anaemia, 38–9, 45–56, 61, 92 82, 120 165, 166, 167 end of life, 227 IgA nephropathy, 158 bicarbonate, 10, 165, 183–4 myeloma, 222 paediatric kidney disease, biochemical tests, 9–13, 10 paediatric patients, 167, 165, 166, 167 bisphosphonates, 151, 219, 219, 167–9, 168 anti-metabolites, 111, 112–13 222

295 296 Index

BK virus, 118–19 peritoneal dialysis, 97, 98, continuous veno-venous blood pressure (BP), 77, 78, 165 103, 174 haemofiltration (CVVHF), 94, blood transfusions, 49, 52, 222 cephalosporins, 101, 102 95, 135, 135, 182 body mass index (BMI), 249 cetirizine, 234 contrast media, 26 body surface area (BSA), 131–2, chemotherapy, myeloma, 220–1 controlled drugs, travel with, 258 164, 178 chlorambucil, 159 co-proxamol, 196 bone pain, myeloma, 219 chloroquine, 260, 261 coronary heart disease (CHD) bortezomib, 221 chlorpheniramine, 234 hyperlipidaemia, 81, 83 buffers, renal replacement fluid, chlorpropamide, 210 hypertension and, 77, 78, 79 183–4 cholesterol, total (TC), 81, 83 corticosteroids bumetanide, 228 chronic allograft nephropathy autoimmune disease, 150, buprenorphine, 197, 230, 231 (CAN), 107, 109, 116 150–1, 154, 155, 156, 158 chronic kidney disease (CKD) myeloma, 221–2 calcification, extraskeletal, 61–3, stage 5 management, 85, renal transplant recipients, 62, 69–70 226–33 107, 111, 112, 115 calcimimetics, 71 stages, 17, 17, 35, 36 CosmoFer, 53 calcineurin inhibitors, 111, 113, chronic renal failure (CRF), 35–44 co-trimoxazole, 119, 153 113–14 causes, 37, 37–8 COX-2 inhibitors, 192, 193, 194–5 adverse effects, 140–1 clinical features, 35, 38–41 cramps, 91 drug interactions, 117, 117 effects on drug handling, creatinine, serum, 9, 10, 16, 17, 21 calciphylaxis (calcific uraemic 41–2 diabetes mellitus, 207 arteriolopathy), 61–2 vs acute renal failure, 28 paediatric patients, 163 calcitriol (1α,25- Churg–Strauss syndrome, 148, creatinine clearance (CrCl), 18, dihydroxycholecalciferol), 7, 149, 149 130, 131 57, 58, 66 ciclosporin, 107, 113, 113–14 renal replacement therapy, calcium, 13, 58 adverse effects, 82, 140–1 100, 135, 135 paediatric kidney disease, autoimmune disease, 159 critical care, 181–9 165–7 generic, 114 cryoglobulinaemic renal bone disease, 39, 40, 58, interactions, 83, 117, 117 glomerulonephritis, 159–60 59, 61 cinacalcet, 71 crystalloid infusions, 30 serum, 10, 13, 58, 69 cinnarizine, 257 crystalluria, 142–3 supplements, 68 ciprofloxacin, 174, 175 cyclophosphamide calcium acetate, 63–4, 166 clonazepam, 40, 234 autoimmune disease, 150, calcium carbonate, 63–4, 68, 166 clonidine, 92 151, 154, 156, 159 calcium channel blockers, 79, 80, co-careldopa, 234 myeloma, 221 80, 81 Cockroft and Gault equation, 18, cystatin C, 130 paediatric kidney disease, 131 cytomegalovirus (CMV), 118 165, 166, 167 codeine, 192, 196, 197, 230, 231 calcium gluconate, 12, 31 collecting system, 2, 5 daclizumab, 110–11 calcium resonium, 12, 31 colloid infusions, 30 dalteparin, 188 calcium–phosphate product, 61, constipation, 226–7, 227, 255 dapsone, 153 62, 69 continuous ambulatory peritoneal darbopoetin (Aranesp), 52–3, 54, capsaicin, 234 dialysis (CAPD), 98, 100, 174 222 captopril, 165, 167 continuous arterio-venous deep vein thrombosis, 257 carbamazepine, 40, 201 haemodiafiltration (CAVHDF), DEET, 256 carbohydrate (CHO), dietary, 240, 94, 135, 135 delayed graft function (DGF), 108, 246 continuous arterio-venous 121 cardiovascular disease, 41 haemodialysis (CAVHD), desmopressin (DDAVP), 15, 15 anaemia as risk factor, 48–9 94 dexamethasone, 221, 226 calcification and, 62–3, 69–70 continuous arterio-venous dextropropoxyphene, 196, 197 diabetic nephropathy, 207 haemofiltration (CAVHF), 94, diabetes mellitus, 82, 205–16 hypertension and, 38, 77, 135, 135 peritoneal dialysis, 96 78–9 continuous veno-venous transplantation, 107–8, 108, NSAID-related risk, 194–6 haemodiafiltration (CVVHDF), 213 carnitine, 91 94, 95, 135, 135, 182 diabetic neuropathy, 199, 200, 201 catheters continuous veno-venous diabetic renal disease, 205–16 central venous, 87, 95, 176 haemodialysis (CVVHD), 94, 95 definitions, 205–6 Index 297

diabetes management, end-of-life care, 233–5 cryoglobulinaemic, 159–60 207–13 energy intake, 246 drug-induced, 141, 141 diagnosis, 206 pre-dialysis, 240, 240–1 membranoproliferative, 159 pathological changes, 206, renal replacement therapy, membranous, 158–9 206 244, 245 rapidly progressive (RPGN), prevention, 206–7 enoxaparin, 118, 188 147 screening and referral, 207 enoximone, 188 glomerulus, 2, 3, 3–4, 36 dialysers, 88–9, 95 enteral feeding, 188, 249–51 glucagon-like peptide 1 analogue, dialysis, 85, 86, 173–4 enterococcal infections, 102 213 management without, 225–37 epoetin alfa (Eprex), 52–3, 54 Goodpasture’s syndrome, 145, see also haemodialysis; epoetin beta (Neo-Recormon), 150, 153–4 peritoneal dialysis 52–3, 54, 222 graft rejection, 108–9 diamorphine, 197, 231, 232–3 epoprostenol, 90, 94–5, 184 acute, 109, 110, 115–16 diclofenac, 117, 193, 195 erythropoiesis, 45, 47–8, 48 chronic see chronic allograft dietary management, 239–52, 246 erythropoiesis-stimulating agents nephropathy acute renal failure, 31, 245–7 (ESAs), 49, 51–4, 222 hyperacute, 108–9 paediatric kidney disease, erythropoietin, 6–7, 38, 45 treatment, 115–16 164–5 inadequate synthesis, 48, 48 grafts, polytetrafluoroethylene pre-dialysis, 41, 65, 239–44, treatment, 39, 52–3, 120, 167, (PTFE), 87–8 246 168 Gram-negative infections, 102, renal replacement therapy, everolimus, 114 175 244–7, 246 excretion, renal, 7, 41 grapefruit juice, 117 type 2 diabetes, 208 exercise, 79, 208 growth, paediatric kidney disease, dihydrocodeine, 196, 197, 230, ezetimibe, 83, 120 164–5 231 α 1 ,25-dihydroxycholecalciferol see fat, dietary intake, 240, 246 H2-antagonists, 187–8 calcitriol fentanyl, 185–6, 187, 197, 198, haematological tests, 13, 13 disequilibrium syndrome, 89, 90, 230–2, 231 haematuria, 147, 157 92, 97–8 ferritin, serum, 10, 39, 46, 51, 168, haemodiafiltration, 95–6 distal convoluted tubule, 2, 4, 5 178 haemodialysis, 85–91 distribution, drug, 128–9, 133–4 fibrates, 83, 120 anticoagulation, 90 diuresis, alkaline, 182 fistula, arterio-venous, 86, 87, 176 complications, 91–2 domperidone, 165, 229 flucloxacillin, 97, 100, 174 continuous, 94, 182–4 dopamine, 29–30 fluconazole, 177 dialysate, 89–90 doses fludrocortisone, 91 dietary management, 244, loading, 132, 136 fluid management, acute renal 246 maintenance, 132 failure, 28–9 drug dosing, 133–4, 134, 135, dosing, drug, 127–37 fluid restriction, 90, 176–8, 242–4, 135 critically ill patients, 188 245, 246 duration and adequacy, 90–1 paediatric kidney disease, 178 folic acid, 169, 169, 261 insulin therapy, 213 renal impairment, 41–2, fungal infections, 102, 118, 177 paediatric patients, 176 127–33 furosemide, 29, 166, 228 parenteral nutrition during, renal replacement therapy, 251 133–6, 134, 184 gabapentin, 192, 200, 201, 234 process, 88, 88–9 doxycycline, 260, 261 gelatin solutions, 30 travelling, 253–4 drug-induced kidney disease, 26–7, gentamicin, 128, 174 vascular access, 86–8 139–44 Giovanetti diet, 239 haemofiltration, 91–5, 93 dyslipidaemia, 81–3 glomerular filtration rate (GFR), 3, critical care, 182–4 dyspnoea, 227–8 16–17, 17 drug dosing, 133, 134, 134–5 childhood estimation, 163–4, haemoglobin, 45 echinacea, 117 164 concentration, 13, 13, 39, electron beam computed chronic kidney disease, 17, 50–1, 53 tomography (EBCT), 70 35, 36 haemodiafiltration and, 96 electrophoresis, serum and urine, estimation, 18, 131–2, 207 paediatric kidney disease, 15–16 measurement, 17–18, 129–31 167, 167–8, 168 elimination, drug, 41, 129 glomerulonephritis, 37, 37, 145 glycated (HbA1c), 207, 208, enalapril, 165, 166 chronic, 147 208 298 Index hair loss, 92 infections local anaesthetics, 192 haloperidol, 40, 229 dialysis patients, 87, 92, 97, loop diuretics, 29–30, 81, 140, 228 headache, dialysis-related, 90, 92 103 loop of Henle, 2, 4, 4–5 Henoch–Schönlein purpura, 157, myeloma, 222 loperamide, 256 158 renal transplant recipients, lorazepam, 186, 187 heparin, 90, 94–5, 100, 103, 184 118–19 low-molecular-weight heparin, 90, see also low-molecular-weight see also peritonitis 95, 118, 188 heparin infliximab, 152, 156 lupus nephritis, 150, 154, 155, hepatitis B vaccination, 169, 170, influenza vaccination, 170, 173 155, 156 171, 262 inotropic therapy, 188 hepatitis C infection, 160 insect bites, 256–7 magnesium, 10, 247 hernia, 104 insulin, 120 malaria, 259–61, 260 holidays, 253–65 analogues, 212, 213 Malarone, 260, 260, 261 hormones, 6–7 intravenous (IV), 12, 31 malignant disease, 119–21 human leukocyte antigens (HLAs), oral (Exubera), 213 malnutrition, 104, 248–51 108–9 therapy in diabetes, 208, mannitol, 29–30 hydromorphone, 197, 198, 231, 212–13 measles, mumps and rubella 232 intensive care units, 181–9 (MMR) vaccine, 169, 170, hydroxychloroquine, 155–6 intravenous excretory urogram 171–2 hyoscine, 234, 257 (IVU), 16 medroxyprogesterone, 226 hypercalcaemia, 61, 66, 219, 222 inulin, 130 mefloquine, 260 hyperkalaemia, 11, 11–12, 241 investigations, 9–19 meglitinides, 211 acute renal failure, 27, 31 iohexol, 131 melphalan, 220 paediatric kidney disease, 169 iron, 10, 45–6, 47 membrane stabilisers, 201 hyperlipidaemia, 81–3, 245 deficiency, 46, 48, 51 mesna, 151 hyperparathyroidism (HPT), 66–7 dietary intake, 46, 247 metabolism, drug, 41, 129 secondary, 39, 59, 59, 61 paediatric kidney disease, metformin, 208, 209, 209–10, 212 tertiary, 61, 66 167–8, 168, 178, 178 methadone, 197, 198, 231, 232 hyperphosphataemia, 39, 57, 61, storage, 46, 51, 51 methotrexate, 151–2 66, 241 therapy, 39, 53–4, 120, 167–8, methylprednisolone, 115, 150, management, 63–5, 69–70 168 150–1, 156, 159 hypertension, 38, 77–81 iron dextran, 53 metoclopramide, 40, 229 see also antihypertensive iron sucrose, 53 metolazone, 81, 228 drugs ischaemic heart disease (IHD) see mexilitene, 201 hypokalaemia, 11, 11–12 coronary heart disease microalbuminuria, 14, 15, 205 hypotension, 91, 181–2 islet cell transplantation, 213 microscopic angiitis, 148, 149, hypovolaemia, 181–2 isotopes, radioactive, 130 149, 152 acute renal failure, 22–3, 28, midazolam, 186, 187, 228, 233 30 ketamine, 192, 199 midodrine, 91 drug-induced, 140 kidneys, 1–7, 2 miglitol, 209 migraine, dialysis-related, 92 ibuprofen, 195, 196 laboratory tests, 9–19 milrinone, 188 icodextrin, 98, 99, 100 lactate, renal replacement fluid, minerals, 247 IgA nephropathy, 156–8 183–4 Modified Diet in Renal Disease immunisation, childhood, 169, lamotrigine, 201 (MDRD) equation, 18, 131–2, 170, 171–3 lansoprazole, 229 207 immunoglobulins, intravenous, lanthanum carbonate, 65, 70 morphine, 185, 187, 196, 197, 152–3 laxatives, 140, 226–7, 227 231, 232–3 immunological tests, 15 left ventricular hypertrophy (LVH), moxonidine, 81 immunosuppressive drugs 38, 48–9, 77 mTOR inhibitors, 112, 114–15, autoimmune disease, 150, lenalidomide, 221 117 150–2, 153–4, 156, 158, 159 levomepromazine, 229 mupirocin, nasal, 92, 97, 102, 177 interactions, 116–17, 117 lidocaine, 201 muromonab CD3 (OKT3), 115–16 paediatric renal transplants, lifestyle modification, 78, 79, 83, muscle relaxants, 186–7 173 208 mycophenolate renal transplantation, 107, light chain deposition disease, 219 autoimmune disease, 150, 109–17 light chains, 217, 218–19 152, 156 Index 299

renal transplant recipients, pain pioglitazone, 209, 210–11 107, 112–13, 116, 173 assessment, 191 plasma exchange, 152, 153–4, myeloma, 217, 217–24 control, 191–204, 229–33 221–2 kidney, 217, 218–19, 222 palliative care, 225–37 pneumococcal vaccine, 169, 170, treatment, 219–22 pamidronate, 219, 219 172–3 myeloperoxidase (MPO), 15, 148 pancreas transplantation, 107–8, Pneumocystis carinii pneumonia, 108, 213 119 naproxen, 193, 195 paracetamol, 192–3, 230, 231 polycystic kidney disease (PKD), nateglinide, 209, 211 paraproteins, 217 37, 37–8 nausea and vomiting, 40, 90, 92 parathyroid adenomas, 67, 71 post-transplant paediatric kidney disease, 165 parathyroid hormone (PTH), 58–9, lymphoproliferative disorders palliative care, 228–9, 229 59, 69, 165–7 (PTLDs), 119–21 nephritic syndrome, 147, 155 parathyroidectomy, 66–8 potassium, 10, 10–12, 169 nephron, 2–3, 3 parenteral nutrition, 251 dietary intake, 241, 243, 244, nephrotic syndrome, 25, 82, 147 paricalcitol, 70–1 245, 246 N-methyl-D-aspartate receptor pergolide, 234 renal replacement fluid, 183 (NMDA) antagonists, 199 peritoneal dialysis, 96–103 prednisolone non-steroidal anti-inflammatory access, 97, 97–8 autoimmune disease, 150, drugs (NSAIDs), 192, 193–6 adequacy, 100–1, 101 150, 154, 156, 158, 159 adverse effects, 26, 140, automated (APD), 98–100, renal transplant recipients, 193–4, 195–6 174 103, 112 palliative care patients, 230 complications, 101–3, 103–4 pregabalin, 201 renal transplantation, 117 continuous ambulatory probenicid, 117 systemic lupus (CAPD), 98, 100, 174 proguanil, 260, 261, 261 erythematosus, 155 dietary management, 244–5, promethazine, 257 nutritional status, assessment, 248, 246 propofol, 186, 187 248–9 drug dosing, 134, 134, 135, prostaglandins, 193 nutritional supplements, 249, 250 135 protein, dietary intake, 246 nutritional support, 164–5, 249–51 fluids, 98, 99 dialysed patients, 244, 245 nystatin, 175 insulin therapy, 213 pre-dialysis, 239, 240, 240 line break/contamination, protein binding, 128–9, 133 obesity, 209, 240–1, 245, 249 176 proteinuria, 14, 147, 165 obstructive nephropathy, 27, 27, paediatric patients, 174–6 proton pump inhibitors, 188 142–3 tidal, 99–100 proximal convoluted tubule, 2, 4, oedema, 39–40 travelling, 254 4 OKT3, 115–16 peritoneal equilibrium test (PET), pruritus, 40, 61, 91, 233, 234 oliguria, 21, 29, 139 100 Pseudomonas infections, 102, 103 omeprazole, 229 peritonitis, 100–3 pure red cell aplasia (PRCA), 54 ondansetron, 40, 234 paediatric patients, 174–6, pyelonephritis, acute, 26 opioids, 196–9, 197 175, 177 critically ill patients, 184, post-surgical, 176 radiological tests, 16 185–6, 187 sclerosing, 103 radiotherapy, myeloma, 220 palliative care, 228, 229, pethidine, 129, 197, 198 ranitidine, 188, 229 230–3, 231, 234 pharmacodynamics, 129 red blood cells (RBCs), 13, 45, 46 oral hypoglycaemic agents (OHA), phosphate, 57, 58, 63 hypochromic, 50, 51 120, 208–12, 209 dietary restriction, 65, 241, remifentanil, 186, 187 osteitis fibrosa, 60 242, 244, 245, 246 renal angiogram, 16 osteomalacia, 60 paediatric kidney disease, renal artery stenosis (RAS), 81, 140 osteopenia, 60 165–7 renal biopsy, 15 over-the-counter (OTC) medicines, renal bone disease, 39, 40, 58 renal blood flow, 3–4, 22, 139, 139 254–5, 255 serum, 10, 57, 68 renal bone disease overweight, 79, 209, 249 see also hyperphosphataemia (osteodystrophy) (ROD), 39, oxycodone, 191–2, 197, 198, 231, phosphate binders, 63–5, 68, 70, 57–75, 60 233 120 children, 167 paediatric kidney disease, pathophysiology, 40, 57–9, 59 paediatric kidney disease, 163–80, 166, 177–8 renal function 164 pilocarpine, 226 assessment, 16–18, 129–32 300 Index renal function continued sodium bicarbonate, 12, 182 paediatric kidney disease, 165 childhood estimation, 163–4, sodium valproate, 200, 201 plasma, 9–10, 10 164 St John’s wort, 117 urea kinetic modelling (Kt/V), 90, classification, 132 staphylococci, coagulase-negative 91, 100 renal replacement therapy (RRT), (CNS), 102 urea reduction ratio (URR), 90 85–106 Staphylococcus aureus, 102, 103, ureteric obstruction, 27, 142–3 acute renal failure, 30–1, 177 urinary tract infections (UTI), 118 182–4 statins, 41, 82, 83, 117, 120 urine output, 21, 29, 139 continuous (CRRT), 91–5, stress ulceration prophylaxis, 31, urine tests, 13–14 135, 182–4 41, 187–8 urokinase, 91, 103 dietary management, 244–7, stroke, 77, 78–9 246 sucralfate, 187–8 vaccinations, 116 drug dosing, 132–6, 184 sulphonylureas, 209, 210, 212 paediatric patients, 169, 170, insulin therapy, 213 sun protection, 254, 257 171–3 paediatric patients, 173–8 symptom management, palliative, travel, 261–3 renal transplantation, 107–25, 226–33 vancomycin, 95, 97, 100–1, 174, 108, 120 systemic lupus erythematosus 175 dietary management, 245 (SLE), 154–6, 160 varicella vaccination, 169, 170, paediatric patients, 169–73 172 travelling, 254, 257 tacrolimus, 107, 112, 113, 113–14 vasculitis, 148, 149 see also graft rejection adverse effects, 83, 113, vecuronium, 186–7, 187 renin, 6 140–1 venlafaxine, 199, 200 repaglinide, 209, 211 interactions, 116, 117, 117 Venofer, 53 restless legs syndrome, 40, 233, tamoxifen, 103 vitamin D, 7, 57, 58 234 teicoplanin, 176 metabolism, 40, 58, 129 retroperitoneal fibrosis, 142 thalidomide, 221, 226, 234 renal bone disease, 39, 40, 58, rituximab, 111, 152, 156 therapeutic drug monitoring 60 rosiglitazone, 209, 210–11 (TDM), 128–9 treatment, 65–6, 70–1, 120, thiazide diuretics, 80, 80, 81, 82 166–7 salbutamol, 12, 31 thiazolidinediones (TZDs), 208, vitamins, 247–8 sedation, critically ill patients, 209, 210–11 volume depletion see 184–7 thromboprophylaxis, 188, 257 hypovolaemia selective serotonin reuptake thrombosis, 86, 91, 118 volume of distribution (Vd), 128, inhibitors (SSRIs), 199, 200 tramadol, 197, 198, 230, 231 129, 133–4 sertraline, 91 transferrin saturation, 46, 50, 51 vomiting see nausea and vomiting sevelamer, 64–5, 70, 166 travel sickness, 257 sieving coefficient, 135 traveller’s diarrhoea, 255, 255–6 warfarin, 87 sirolimus, 107, 112, 114–15, 173 travelling, 253–65 Wegener’s granulomatosis, 148–9, adverse effects, 82–3, 114 tricyclic antidepressants (TCAs), 149, 152 interactions, 117, 117 192, 199, 199–200 weight, body, 79, 90 slow continuous ultrafiltration trimethoprim, 118 ideal, 131 (SCUF), 94 tuberculosis (TB), 102, 119 smoking, 79, 83, 153 X-rays, 16 sodium, 10, 12–13 ultrafiltration profiling, 90, 91 dietary intake, 79, 165, ultrasound, 16 yellow fever vaccination, 262–3 242–4, 245, 246 urea, 7, 130 profiling, 90, 91 clearance rate, 184 zinc, 92, 226