You are on page 1of 98

Competency-Based Critical Care

Series Editors
John Knighton, MBBS, MRCP, FRCA Paul Sadler, MBChB, FRCA
Consultant Consultant
Intensive Care Medicine & Anaesthesia Critical Care Medicine & Anaesthesia
Portsmouth Hospitals NHS Trust Queen Alexandra Hospital
Portsmouth Portsmouth
UK UK

Founding Editor
John SP Lumley
Emeritus Professor of Vascular Surgery
University of London
London
UK
and
Honorary Consultant Surgeon
Great Ormond Street Hospital for Children NHS Trust (GOSH)
London
UK

Other titles in this series


Sepsis
Simon Baudouin (Ed.)
Sara Blakeley (Ed.)

Renal Failure and


Replacement
Therapies
Sara Blakeley, BM, MRCP, EDIC
Queen Alexandra Hospital
Portsmouth
Hampshire, UK

British Library Cataloguing in Publication Data


Renal Failure and Replacement Therapies.—(Competency-based critical care)
1. Kidneys—Diseases 2. Kidneys—Diseases—Treatment
3. Acute renal failure 4. Acute renal failure—Treatment
I. Blakeley, Sara
616.6′1
ISBN-13: 9781846289361

Library of Congress Control Number: 2007927934

Competency-Based Critical Care Series ISSN 1864-9998

ISBN: 978-1-84628-936-1 e-ISBN: 978-1-84628-937-8

© Springer-Verlag London Limited 2008

Apart from any fair dealing for the purposes of research or private study, or criticism or review, as
permitted under the Copyright, Designs and Patents Act 1988, this publication may only be repro-
duced, stored or transmitted, in any form or by any means, with the prior permission in writing
of the publishers, or in the case of reprographic reproduction in accordance with the terms of
licences issued by the Copyright Licensing Agency. Enquiries concerning reproduction outside
those terms should be sent to the publishers.
The use of registered names, trademarks, etc. in this publication does not imply, even in the absence
of a specific
fi statement, that such names are exempt from the relevant laws and regulations and
therefore free for general use.
Product liability: The publisher can give no guarantee for information about drug dosage and
application thereof contained in this book. In every individual case the respective user must check
its accuracy by consulting other pharmaceutical literature.

9 8 7 6 5 4 3 2 1

springer.com
Contents

Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vii

Chapter 1 Assessment of Renal Function . . . . . . . . . . . . . . . . . . . . . 1


Mohan Arkanath

Chapter 2 Imaging of Acute Renal Failure—A Problem-Solving


Approach for Intensive Care Unit Physicians . . . . . . . . 7
Tom Sutherland

Chapter 3 Drug-Induced Renal Injury . . . . . . . . . . . . . . . . . . . . . . . . 14


Sara Blakeley

Chapter 4 Acute Kidney Injury . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19


Sara Blakeley

Chapter 5 Medical Management of Acute Renal Failure . . . . . . . . 26


Nerina Harley

Chapter 6 Acute Renal Failure in the Surgical Patient . . . . . . . . . . 33


Marlies Ostermann

Chapter 7 Rhabdomyolysis and Compartment Syndrome . . . . . . . 38


Laurie Tomlinson and Stephen Holt

Chapter 8 Multisystem Causes of Acute Renal Failure . . . . . . . . . . 42


Tim Leach

Chapter 9 Therapeutic Plasma Exchange . . . . . . . . . . . . . . . . . . . . . 49


Tim Leach

Chapter 10 Renal Replacement Therapy . . . . . . . . . . . . . . . . . . . . . . . 51


John H. Reeves

Chapter 11 Technical Aspects of Renal Replacement Therapy . . . . 57


Sara Blakeley

v
vi Contents

Chapter 12 End-Stage Renal Disease . . . . . . . . . . . . . . . . . . . . . . . . . . 64


Emile Mohammed

Chapter 13 Clinical Hyperkalemia and Hypokalemia . . . . . . . . . . . . 71


Harn-Yih Ong

Chapter 14 Clinical Hyponatremia and Hypernatremia . . . . . . . . . . 77


Himangsu Gangopadhyay

Chapter 15 Clinical Metabolic Acidosis and Alkalosis . . . . . . . . . . . 81


Sara Blakeley

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
Contributors

Mohan Arkanath, MRCP School of Clinical Medicine and Research


Consultant Nephrologist Queen Elizabeth Hospital & Cavehill Campus
Doncaster Royal Infi
firmary Bridgetown, Barbados
Doncaster, UK
Harn-Yih Ong
Sara Blakeley, BM, MRCP, EDIC Registrar Intensive Care
Queen Alexandra Hospital St Vincent’s Hospital
Portsmouth Melbourne, Victoria
Hampshire, UK Australia
Nerina Harley, MBBS, MD, PGDIPEcho,
FRACP, FJFICM Marlies Ostermann
Intensive Care Consultant Consultant Nephrology and Critical Care
The Royal Melbourne Hospital Intensive Care Unit
Victoria, Australia St Thomas’ Hospital
London, UK
Stephen Holt, PhD, FRCP
Consultant Nephrologist and Honorary John H. Reeves, MD, MBBS, FANZCA, FJFICM,
Senior Lecturer EDIC
Sussex Kidney Unit Consultant in Intensive Care and Anaesthetics
Brighton and Sussex University Hospitals Department of Anaesthesia and Pain
Royal Sussex County Hospital Management
Brighton, UK Alfred Hospital
Melbourne, Victoria
Himangsu Gangopadhyay, MD, MBBS, Australia
FFARCS(Ireland), FJFICM
Consultant in Intensive Care
Tom Sutherland, MBBS (Hons)
Frankston Hospital
Radiology Registrar
Frankston, Victoria, Australia
St Vincent’s Hospital
Tim Leach, BM, FRCP Melbourne, Victoria
Consultant Nephrologist Australia
Wessex Renal and Transplant Unit
Queen Alexandra Hospital Laurie Tomlinson, MRCP
Portsmouth, UK Specialist Registrar Nephrology
Sussex Kidney Unit
Emile Mohammed, MB, ChB, MRCP (UK) Brighton and Sussex University Hospitals
Lecturer in Medicine (University of the West Royal Sussex County Hospital
Indies) and Consultant Nephrologist Brighton, UK

vii
1
Assessment of Renal Function
Mohan Arkanath

Normal Functions of the Kidney sure are dependent on optimum plasma volume,
and most enzymes function best in narrow ranges
To be able to assess a degree of renal function or of pH or ion concentration.
dysfunction, it is important to first
fi consider the The kidneys take up the role of correcting any
normal functions of the kidney: alterations in the composition and volume of body
fluids that occur as a consequence of food intake,

1. Maintenance of body composition: The metabolism, environmental factors, and exercise.
volume of fl fluid in the body, its osmolarity, elec- In healthy individuals, these corrections occur in
trolyte content, concentration, and acidity are all a matter of hours, and body fluid
fl volume and the
regulated by the kidney via variation in urine concentration of most ions return to normal set
excretion of water and ions. Electrolytes regulated points. In many disease states, however, these
by changes in urinary excretion include sodium, regulatory processes are disturbed, resulting in
potassium, chloride, calcium, magnesium, and persistent deviations in body fl fluid volume and
phosphate. composition.
2. Excretion of metabolic end products and
foreign substances: The most notable are urea and
a number of toxins and drugs. Body Fluid Composition
3. Production and secretion of enzymes and A large proportion of the human body is com-
hormones: posed of water (Table 1.1). Adipose tissue is low in
a. Renin: See Figure 1.1. water content and, hence, obese individuals have
b. Erythropoietin: A glycosylated, 165-amino a lower body water fraction than lean individuals.
acid protein produced by renal cortical Because of slightly greater fat content, women
interstitial cells that stimulates maturation generally contain less water than men.
of erythrocytes in the bone marrow.
c. 1, 25-Dihydroxyvitamin D3: The most active
form of vitamin D3, it is formed by proximal Clinical Evaluation of Renal Function
tubule cells. This steroid hormone plays an
important role in the regulation of body Glomerular filtration
fi rate (GFR) is generally con-
calcium and phosphate balance. sidered the best measure of renal function; it is the
sum of filtration
fi rates of all of the functioning
nephrons. It is defined
fi as the renal clearance of a
The Role of the Kidney in Homeostasis particular substance from plasma, and is expressed
Maintenance of body fluid fl composition and as the volume of plasma that can be completely
volume are important for many functions of the cleared of that substance in a unit of time. In the
body. For example, cardiac output and blood pres- following sections, we will compare the advantages

1
2 M. Arkanath

Renin TABLE 1.2. Other factors altering blood urea or serum creatininea
Angiotensinogen Angiotensin Salt balance Increased level Decreased level
(Plasma globulin) (Vasoconstrictor) and blood
Urea Prerenal causes (e.g., Cirrhosis
pressure
control congestive heart failure, Protein malnutrition
volume contraction) lead Water excess (e.g.,
FIGURE 1.1. Production and secretion of enzymes and hormones to increased tubular SIADH, saline
for renin. reabsorption infusion) leads to
• Gastrointestinal bleeding reduced tubular
• Catabolic state reabsorption
• Corticosteroids
• Hyperalimentation
and disadvantages of the various methods avail- • Tetracyclines
able for GFR estimation or quantification.
fi Creatinine Overproduction (e.g., Decreased muscle
rhabdomyolysis, vigorous mass
Clearance Urine mg/dL Volume mL/min ) sustained exercise,
= anabolic steroids, dietary
(mL/min
/ i ) Plasma mg/dL ) supplements such as
creatine)
Where x is the substance being cleared. Blocked tubular secretion
Features of an ideal marker for GFR (e.g., drugs such as
trimethoprim and
determination:
cimetidine)
• Appears endogenously in the plasma at a con- Assay interference (e.g.,
ketosis and drugs such as
stant rate
cephalosporins, flucytosine,
• Is freely filtered by the glomerulus methyldopa, levodopa,
• Does not undergo reabsorption or secretion by and ascorbic acid)
the renal tubule a
SIADH, syndrome of inappropriate ADH secretion.
• Is not eliminated by extrarenal routes

Urea
Creatinine
Urea was first isolated in 1773, and urea clearance
as a surrogate marker for GFR introduced in 1929. Creatinine is a metabolite of creatine and phos-
Although urea measurement is performed fre- phocreatine found in skeletal muscle. It is a
quently, it is well recognized that its many draw- small molecule that is not protein bound and,
backs make it a poor measure of renal function hence, freely filtered by the glomerulus. It does,
(see Table 1.2). For example, the rate of production however, undergo tubular secretion which is
is influenced
fl by factors such as the availability variable (see Table 1.2). Creatinine production
of nitrogenous substrates, and the rate of reab- can vary in an individual over time with
sorption in the tubules can be affected by volume muscle mass changes or acutely with massive
status. myocyte turnover. There are also age- and sex-
associated differences in serum creatinine (Sα)
concentration; it is lower in the elderly and in
women.
TABLE 1.1. Body fluid compartment volumesa The ratio between serum urea and creatinine
Example for a 60-kg patient can be useful when assessing the patient with
TBW = 60% × body weight 60% × 60 kg = 36 L
acute renal failure. Under normal circumstances,
ICW = 2/3 TBW / × 36 L = 24 L
23 the ratio between urea and creatinine is 10 : 1 but
ECW = 1/3 TBW 1/3× 36 L = 12 L this value can rise to greater than 20 : 1 when the
Plasma water = 1/4 ECW 1/4 × 12 L = 3 L
extracellular volume is contracted. A volume-con-
Blood volume = Plasma water ÷ 3 L ÷ (1 − 0.40) = 6.6 L tracted state ((prerenal) is a “sodium avid” state
(1 − hematocrit)
and promotes proximal tubular and distal nephron
a
TBW, total body water; ICW, intracellular water; ECW, extracellular water. reabsorption of urea but not creatinine. Acute
1. Assessment of Renal Function 3

tubular necrosis (ATN) will have a urea-to-creati- Urinalysis (4, 5)


nine ratio of 10 : 1 because tubular reabsorption of
urea is not preferentially increased. The microscopic examination of the urinary sedi-
ment is an indispensable part of the work-up of
patients with renal insuffificiency, proteinuria, hema-
Assessment of GFR turia, urinary tract infections, or kidney stones. A
Sα has become a standard measure of renal func- careful urinalysis has been referred to as a “poor
tion because of its convenience and low costs; man’s renal biopsy.” The urine should be collected
however, it is crude marker of GFR. A 24-hour as a midstream catch or fresh catheter specimen,
creatinine clearance (Cα) is often used in practice and because the urine sediment can degenerate
as a measure of the GFR because creatinine is with time, it should be examined soon after collec-
freely fi
filtered and not reabsorbed by the tubule. tion. Urinalysis should include a dipstick examina-
However, approximately 15% of urinary creati- tion for specifi fic gravity, pH, protein, hemoglobin,
nine is a result of tubular secretion and, thus, this glucose, ketones, nitrites, and leucocytes. This
method overestimates GFR. The other problems should be followed by microscopic examination if
with Cα are incomplete urine collection and there are positive fi findings. Microscopic examina-
increasing creatinine secretion. Inulin clearance tion should check for all formed elements: crystals,
is traditionally considered the “gold standard” cells, casts, and infecting organisms.
for the measurement of GFR (1). It is one of the
most accurate measures of renal function, but Appearance
the inconvenience of administration, cost, and
Normal urine is clear, with a faint yellow tinge
limited supply of inulin preclude its use in routine
caused by the presence of urochromes. As it be-
practice.
comes more concentrated, its color deepens. Biliru-
bin, other pathologic metabolites, and a variety of
drugs may discolor the urine or change its smell.
Estimated Ca: The Cockroft and Gault
Formula (2)
Specific Gravity
This formula is used to estimate Cα at the bedside
using age of the patient and Sα value (both corre- The urine specificfi gravity is a conveniently deter-
late inversely with GFR), and the ideal body weight mined but inaccurate surrogate of osmolality. Spe-
(IBW) of the patient. cific
fi gravities of 1.001 to 1.035 correspond to an
This formula can used only when the Sα value osmolality range of 50 to 1000 mOsm/kg. A spe-
is in a steady state and not when it is rapidly cific
fi gravity near 1.010 connotes isosthenuria
changing, as in acute renal failure. It has also been (urine osmolality matching plasma).
shown that Sα and estimations of GFR using The specific fi gravity is used to determine
various formulae are often inaccurate in critically whether the urine is, or can be, concentrated.
ill patients (3). Although they may give an esti- During a solute diuresis accompanying hypergly-
mate, their limitations should be remembered cemia, diuretic therapy, or relief of obstruction,
when assessing renal function in patients on the the urine is isosthenuric. In contrast, with a water
intensive care unit. Other factors, such as urine diuresis caused by overhydration or diabetes
output, clearance of acid and electrolytes, and rate insipidus, the specifi
fic gravity is low. It is also useful
of change of serum urea and creatinine should all in differentiating between prerenal cause of renal
be considered together. failure (high) and ATN.
The Cockroft and Gault formula is:
Volume
(140 − patient age weight in kg )
Estimated C α = In health, the volume of urine passed is primarily
(Sα in μmol/L )
determined by diet and fluid
fl intake. The mini-
Multiply by 0.85 for women, 1.23 if male. mum amount passed to stay in fluid fl balance is
4 M. Arkanath

determined by the amount of solute being excreted TABLE 1.3. Causes of hematuriaa
(mainly urea and electrolytes), and the concen- Glomerular disease Mesangial IgA nephropathy
trating ability of the kidneys. In disease, impair- Thin basement membrane disease
ment of concentrating ability requires increased Mesangial proliferative GN
volumes of urine to be passed for the same solute Membranoproliferative GN
Crescentic GN
output.
Systemic lupus erythematosus
Post streptococcal GN
• Oliguria is defifined as the excretion of less than Infective endocarditis
300 mL/d of urine, and is often caused by intrin- Alport’s syndrome
sic renal disease or obstructive uropathy. Vascular and Acute hypersensitivity interstitial nephritis
tubulointerstitial Tumors (renal cell carcinoma, Wilm’s tumor,
• Anuria suggests urinary tract obstruction until
disease leukemia)
proven otherwise. Polycystic kidney disease
• Polyuria is a persistent, large increase in urine Malignant hypertension
output, usually associated with nocturia. Poly- Analgesic nephropathy
uria is a result of excessive intake of water (e.g., Diabetes mellitus
Obstructive uropathy
compulsive water drinking), increased excretion
Urinary tract diseases Carcinoma (renal pelvis, ureter, bladder,
of solute (hyperglycemia and glycosuria), a urethra, prostate)
defective renal concentrating ability, or failure Calculi
of production of antidiuretic hormone (ADH). Retroperitoneal fibrosis
Tuberculosis
Cystitis
Chemical Testing Drugs (e.g., cyclophosphamide)
Trauma
Blood Benign prostatic hypertrophy
Urethritis
Hematuria may be macroscopic or microscopic Platelet defects (e.g., idiopathic or drug
(Table 1.3). Currently used dipstick tests for blood induced thrombocytopenic purpura)
are very sensitive, being positive if two or more a
GN, glomerulonephritis.
red cells are visible under the high-power fi field
(HPF) of a light microscope. A disadvantage is that
dipstick testing cannot distinguish between blood
and free hemoglobin. A positive dipstick has to be
concentration of 150 mg/L or more in urine. They
followed by microscopy of fresh urine to confirm fi
react primarily to albumin and are insensitive to
the presence of red cells and to exclude rare condi-
globulin or Bence-Jones protein. False-positive
tions such as hemoglobinuria and myoglobinuria.
results are common with iodinated contrast
In female patients, it is essential to enquire whether
agents; hence, urine testing should be repeated
the patient is menstruating.
after 24 hours. The normal rate of excretion of
Abnormal numbers of erythrocytes in the urine
protein in urine is 80 ± 24 mg/24 h in healthy indi-
may arise from anywhere from the glomerular
viduals, but protein excretion rates are somewhat
capillaries to the tip of the distal urethra. Dysmor-
higher in children, adolescents, and in pregnancy.
phic erythrocytes tend strongly to be associated
Fever, severe exercise, and the acute infusion of
with a glomerular source. Abnormal proteinuria
hyperoncotic solutions or certain pressor agents
along with dysmorphic erythrocytes is a reliable
(e.g., angiotensin II or norepinephrine) may
sign of glomerular disease. Urinary tract abnor-
transiently cause abnormal protein excretion
malities lead to microscopic or macroscopic
in normal individuals.
hematuria but the erythrocytes exhibit normal
The protein in normal and abnormal urine is
morphology.
derived from three sources:
Protein
1. Plasma proteins filtered
fi at the glomerulus and
Proteinuria is one the most common signs of renal escaping proximal tubular reabsorption.
disease (Table 1.4). Most reagent strips detect a 2. Proteins normally secreted by renal tubules.
1. Assessment of Renal Function 5

TABLE 1.4. Causes of proteinuria based on pathophysiologic mechanisma


Glomerular proteinuria Primary glomerular disease Secondary glomerular disease
• Minimal change disease • Drugs: e.g., mercurials, gold compounds, heroin,
• Mesangial proliferative GN penicillamine, probenecid, captopril, lithium, NSAID
• Focal and segmental glomerulosclerosis (FSGS) • Allergens: bee sting, pollen, milk
• Membranous GN • Infections: bacterial, viral, protozoal, fungal, helminthic
• Mesangiocapillary GN • Neoplastic: solid tumors, leukemia
• Fibrillary GN • Multisystem: SLE, Henoch-Schonlein purpura, amyloidosis
• Crescentic GN • Heredofamilial: diabetes mellitus, congenital nephritic
syndrome, Fabry’s disease, Alport’s syndrome
• Others: febrile proteinuria, postexercise proteinuria, benign
orthostatic proteinuria
Tubular proteinuria Endogenous toxins: light chain damage to
proximal tubular, lysozyme (leukemia)
Exogenous toxins and drugs: mercury, lead,
cadmium, outdated tetracycline
Tubulointerstitial disease SLE
Acute hypersensitivity interstitial nephritis
Acute bacterial pyelonephritis
Obstructive uropathy
Chronic interstitial nephritis
Overflow proteinuria Multiple myeloma
Light chain disease
Amyloidosis
Hemoglobinuria
Myoglobinuria
Certain colonic or pancreatic carcinomas
Tissue proteinuria Acute inflammation of urinary tract
Uroepithelial tumors
a
GN, glomerulonephritis; NSAID, nonsteroidal anti-inflammatory drugs; SLE, systemic lupus erythematosus.

3. Proteins derived from the lower urinary tract Bacteriuria


or leaking into the urine as a result of tissue
Dipsticks detect nitrites produced from the reduc-
injury or inflammation.

tion of urinary nitrate by bacteria and also leuco-
cyte esterase, an enzyme specificfi for neutrophils.
Most patients with persistent proteinuria should
Detection of both nitrites and leucocytes on dip-
undergo a quantitative measurement of protein
stick has a high predictive value for urinary tract
excretion, with a 24-hour urine measurement.
infection.
A protein excretion rate greater than 3.5 g/d
(nephrotic range proteinuria) should prompt
further investigations to ascertain the exact cause Urine Microscopy
of the proteinuria with measurements of urea,
An unspun sample of urine may be examined
creatinine, liver function tests (most impor-
under low- or high-power microscopy, however, a
tantly, serum albumin), and a full blood count.
spun sample is a more accurate. Urine is centri-
A defifinitive diagnosis has to be achieved with a
fuged, the supernatant is discarded, and an aliquot
renal biopsy. Nephrotic syndrome is definedfi as a
of the residue is placed on a glass slide using a
combination of proteinuria in excess of 3 g/d,
Pasteur pipet. All patients suspected of having
hypoalbuminemia, edema, and hyperlipidemia.
renal disease should have urine microscopy.

Glucose White Blood Cells


Renal glycosuria is uncommon and any positive The presence of 10 or more white blood cells per
test requires evaluation of diabetes mellitus. cubic millimeter is abnormal and indicates an
6 M. Arkanath

infl
flammatory reaction within the urinary tract. casts are indicative of stasis in the collecting
Most commonly this represents a urinary tract tubules and are seen in chronic renal failure.
infection but it may also be found with a sterile
sample in patients on antibiotic therapy, or with
References
kidney stones, tubulointerstitial nephritis, papil-
lary necrosis, or tuberculosis. 1. Brown SC, O’Reilly PH. Iohexol clearance for the
determination of glomerular filtration
fi rate in clinical
practice: evidence for a new gold standard. J Urol.
Red Blood Cells 1991; 146(3): 675–679.
As mentioned previously, erythrocytes can find fi 2. Cockroft DW, Gault MH. Prediction of creatinine
their way into the urine from any source between clearance from serum creatinine. Nephron. 1976; 16:
the glomerulus to the urethral meatus. The pres- 31–41.
3. Hoste EA, Damen J, Vanholder RC, et al. Assessment
ence of more than two to three red blood cells per
of renal function in recently admitted critically ill
HPF is usually pathological. Erythrocytes origi-
patients with normal serum creatinine. Nephrol Dial
nating in the renal parenchyma are dysmorphic, Transplant. 2005; 20(4): 747–753.
whereas those originating in the collecting system 4. Henry JB, Lauzon RB, Schaumann GB. Basic exami-
retain their uniform biconcave shape. nation of the urine. Clinical Diagnosis and Manage-
ment by Laboratory Methods. 19th ed. Philadelphia:
Casts Saunders. 1991.
5. Graff L. A Handbook of Routine Urinalysis. Philadel-
Based on their shape and origin, casts are appro- phia: Lippincott. 1983.
priately named. Hyaline casts are found in con-
centrated urine, during febrile illnesses, after
strenuous exercise, and with diuretic therapy. Suggested Reading
They are not indicative of renal disease. Red cell
Davison AM, Cameron S, Grunfeld J-P, et al. Oxford
casts indicate acute glomerulonephritis. White cell
Textbook of Clinical Nephrology. 3rd ed. Oxford:
casts indicate infection or infl
flammation, and are Oxford University Press. 2005.
seen in pyelonephritis and interstitial nephritis. Greenberg A, Cheung AK, Coffman TM, et al. Primer on
Renal tubular casts are found in cases of ATN and Kidney Diseases. 3rd Ed. London: Academic Press.
interstitial nephritis. Coarse granular casts are 2001.
nonspecificfi and represent the degeneration of a Guyton AC. Textbook of Physiology. 8th Ed. New York:
cast with a cellular element. Finally, broad waxy Saunders. 1991.
2
Imaging of Acute Renal Failure—A
Problem-Solving Approach for Intensive
Care Unit Physicians
Tom Sutherland

Acute renal failure (ARF) is a common problem in (MRI) scans analyze renal structure and renal
hospitalized patients. It has a variety of causes, artery calcifification, and dynamic gadolinium-
traditionally divided into prerenal, renal, and enhanced MRI renal studies allow functional
postrenal. A further classification
fi can be made assessment. Other functional studies, such as mer-
into medical and surgical causes, with the later captoacetyltriglycine (MAG3) and diethylene tri-
defined
fi as patients who will benefifit from mechan- amine pentaacetic acid (DTPA) show reduced renal
ical intervention. uptake and delayed excretion of tracer.
Acute tubular necrosis (ATN) is the most A further role of imaging is to determine the
common cause of ARF (approximately 45%) and number of present and functioning kidneys. For
postrenal obstruction accounts for roughly 10% of ARF to occur in previously normal kidneys, the
presentations (1). A variety of imaging modalities underlying cause must be a bilateral process, or
may be used to help diagnose the cause, or, if this else a single functioning kidney must be compro-
is not possible, to differentiate medical from sur- mised (Figure 2.1).
gical causes. ARF in renal transplants will not be
addressed.
Acute Tubular Necrosis
Acute or Chronic Renal Failure? Ultrasound will usually show enlarged kidneys
with a smooth contour caused by interstitial
This is best answered by clinical assessment rather edema, no hydronephrosis, and renal arterial and
than with imaging. Imaging findingsfi in chronic venous flow. The examination of choice in sus-
renal failure are nonspecific fi and essentially char- pected ATN is a MAG3 nuclear medicine study.
acterized by small kidneys. X-rays can show the Scintigraphic examinations in ATN using Tc-
renal outline, calcifi fication, renal bone disease and 99m-MAG3 demonstrate relatively well-preserved
effects of hyperparathyroidism. Ultrasound is an on-time renal perfusion, and delayed tracer
excellent modality for structural imaging because uptake, often with a continuing activity accumula-
it is able to detect reduced renal parenchyma, scar- tion curve. If the activity curve does have a
ring (usually secondary to previous refluxfl nephro- maximum, the time to maximum is delayed (2).
pathy), calcification,
fi and polycystic kidneys. The Excretion of tracer into the collecting system is
echogenicity of the cortex can be assessed with a delayed and reduced, but there is no obstruction
hyperechoic cortex (normal cortex is hypoechoic to drainage of the collecting systems. If excre-
to liver), present in most causes of chronic renal tion and drainage occur, the time from maximal
failure; adult polycystic kidney disease being the activity to half-maximal activity (or another
notable exception. Noncontrast computed tomo- quantitative measure of tracer excretion) is pro-
graphic (CT) and magnetic resonance imaging longed. What underlies this scintigraphic pattern

7
8 T. Sutherland

Liver Renal sinus Liver Renal cortex Dialysis fluid

A B

FIGURE 2.1. A, ultrasound of a normal kidney. Smooth cortex, hypoechoic to liver. B, chronic renal failure with a small irregular kidney
with hyperechoic cortex compared with liver (anechoic area around the liver is peritoneal dialysis fluid).

FIGURE 2.2. MAG3 study showing progressive accumulation of peak as the kidneys are perfused and then activity declines as
tracer in the renal cortex indicating normal perfusion but no excre- tracer is excreted and passes into the bladder. Lt, left; Rt, right;
tion, consistent with ATN. A normal activity curve should initially Bkg, background.
2. Imaging of Acute Renal Failure 9

is parenchymal retention of MAG3 by the remain- Ultrasound is the first-line


fi test for obstruction.
ing viable tubular cells, whereas tubular obstruc- It is radiation free, portable, is not nephrotoxic,
tion prevents drainage of tracer into the collecting and can simultaneously gather other structural
system. The tubular cells continue to take up tracer information. Obstructed kidneys are typically
while they are viable and, thus, the cortical activity normal sized with dilated ureters, renal pelvis, and
can be used to monitor disease progress, with pro- calyceal systems. These urine-filled fi structures
gressive loss of cortical activity being a poor prog- appear as anechoic areas with posterior acoustic
nostic indicator (Figure 2.2). enhancement. Caution is required in interpreta-
If a MAG3 study cannot be performed, ultra- tion because a ureter and renal pelvis may be
sound will demonstrate a cortex of normal echo- dilated without being obstructed (a false positive).
genicity with either a normal or hypoechoic This can occur after previous obstruction that
medulla. The renal arteries can also be interro- leaves a residual “baggy” collecting system, or as
gated for the renal index (RI), which is an objec- an anatomical variant (enlarged extrarenal pelvis).
tive measure of the resistance to renal perfusion. Differentiation can often be made by examining
RI is defifined as (systolic velocity minus diastolic the bladder for ureteric jets, which are the periodic
velocity) divided by systolic velocity, and has expulsion of urine from the ureter into the bladder,
been heavily investigated to determine whether which can be detected by Doppler imaging. If ure-
elevation in RI can differentiate ATN from renal teric jets are present, then there is not a complete
hypoperfusion not yet complicated by ATN. Unfor- obstruction on that side (3). False negatives can
tunately, there have been mixed results and, gener- occur in the hyperacute setting if the renal collect-
ally, RI has inadequate specifi ficity for routine ing system has not had time to dilate, or if the
clinical use. system has spontaneously decompressed by forni-
ceal or renal pelvis rupture (Figure 2.3).
Noncontrast CT scan is the gold standard for
Glomerulonephritis and Acute detecting ureteric calculi (4). The ureters can
Interstitial Nephritis usually be traced between the kidney and bladder,
and a hyperdense stone can be seen at the distal
Clinical history and urinalysis plays a vital role in site of hydroureter. More than 99% of renal calculi
differentiating GN and acute interstitial nephritis are radiopaque on CT scan, however, xanthine
(AIN) with the “gold standard” diagnostic test calculi may be radiolucent and stones associated
being a renal biopsy. The main role of imaging is with indinavir are radiolucent. The obstructed
to detect structural signs of chronic renal disease kidney is typically edematous (i.e., swollen) with
and to exclude other causes of ARF. MAG3 studies
will show poorly functioning kidneys, but no
accumulation pattern and also no obstruction Cortex Renal pelvis Calyx
to drainage. Edema can sometimes be demon-
strated with ultrasound, manifest as hypoechoic
large kidneys. If there is a clinical suspicion to
direct imaging, a careful search may also find fi
other signs of the underlying cause, for example,
pulmonary hemorrhage in Goodpasture’s syn-
drome and pulmonary nodules in Wegener’s
granulomatosis.

Ureteric Obstruction
For obstruction to produce ARF it must be bilat-
eral or affecting a single functioning kidney. This FIGURE 2.3. Ultrasound of a hydronephrotic kidney. Size is normal
is the classical cause of surgical ARF. with dilated renal pelvis and calyces (the anechoic central part).
10 T. Sutherland

perirenal stranding. The administration of con- the degree of obstruction. Once again, it must be
trast is contraindicated in ARF because of its stressed that the only way mechanical obstruction
potential nephrotoxicity. Although not as helpful can produce ARF in previously normal kidneys is
as a contrast-enhanced CT scanning, a non- by affecting a solitary functioning kidney, or by
contrast study can usually detect many extrinsic blocking both kidneys simultaneously.
compressing masses, such as retroperitoneal
tumors, or cervical or colon carcinomas, that
may produce bilateral obstruction. Complications Renal Artery Dissection or Occlusion
related to trauma, such as urinoma or renal pedicle
avulsions, are also visible even without contrast Each kidney is normally supplied by a single renal
(Figure 2.4). artery that arises from the aorta before dividing
Scintigraphic imaging with either Tc-99m- into an average of five segmental end-arterial
MAG3 or Tc-99m-DTPA can detect ureteric branches. Arterial dissection and occlusion is
obstruction by showing a dilated collecting system another surgical cause of ARF. Renal infarction
with delayed drainage of tracer. The collecting may be caused by blunt trauma with avulsion of
system is outlined down to the level of the obstruc- the renal pedicle or penetrating trauma transect-
tion with no or limited tracer draining beyond ing the renal artery. Renal artery dissection may
this. The negative predictive value of nuclear med- be iatrogenic, occurring during endovascular
icine scanning is extremely high, with a normal intervention. Bilateral dissection or exclusion
study virtually excluding obstruction. Of course, from the circulation can occur secondary to an
sufficient
fi tracer must be present in the collecting aortic dissection. Suspicion of a dissection or
system to reach the obstruction point. Reduced occlusion is virtually the only differential diagno-
urine production associated with renal failure sis that warrants the administration of intra-
may deliver so little tracer into the collecting venous CT contrast in the setting of ARF in which
system that obstruction cannot be excluded. The MRI scanning is unavailable. An arterial phase CT
lower the patients glomerular fi filtration rate (GFR), scan highlights the renal artery anatomy, demon-
the higher the probability of having an indetermi- strating the intimal fl
flap of dissection and can also
nate study (5). Further, an enlarged but non- detect signs of renal infarction and lacerations (6)
obstructed collecting system may mimic delayed (Figure 2.5).
drainage. In this latter situation, the specifificity of Magnetic resonance angiography (MRA) has
the examination is increased by administering excellent sensitivity and specifi ficity for detecting
20 mg (or sometimes 40 mg) of frusemide IV, and dissection and occlusion of proximal renal arter-
scanning for a further 20 minutes. A baggy non- ies. Segmental arteries are less well visualized,
obstructed system will promptly wash out, and the however, unilateral or isolated segmental pathol-
delay in washout after frusemide correlates with ogy will not produce ARF. MR contrast is much

Arrow heads are fatty stranding. Right ureteric calculus Inferior pole left kidney

FIGURE 2.4. Noncontrast CT scan with


complete right-sided and moderate left-
sided obstruction secondary to bilateral
ureteric calculi (left calculus not shown).
Edematous fatty stranding around each
kidney with the right calculus in the
dilated ureter. Arrowheads indicate fatty
stranding.
2. Imaging of Acute Renal Failure 11

FIGURE 2.5. Contrast enhanced CT scan Contrast in the aorta left kidney
(magnified view of the left kidney) with
no renal parenchymal enhancement or
renal vascular enhancement secondary
to an occlusive renal artery embolus.

less nephrotoxic than CT contrast. MRI scanning of blood flowing around the thrombus is shown
is logistically difficult
fi for most intensive care unit on color Doppler imaging (7). The kidney itself
(ICU) patients, and has a considerably longer scan may have reduced corticomedullary differentia-
time and, therefore, CT scanning remains a more tion, be enlarged, and contain focal areas of
practical modality. increased echogenicity related to edema and hem-
MAG3 examinations will reveal an absence of orrhage. Contrast-enhanced CT scanning has a
perfusion (segmental or unilateral or bilateral, as similar appearance in the acute setting, with a
the case may be) but not the cause. dilated renal vein with hypodense thrombus
within. Secondary findings,
fi such as perirenal
stranding, dilated gonadal vein, prolonged nephro-
Renal Vein Thrombosis gram, and complications such as retroperitoneal
hemorrhage may also be present. As the chronicity
Bilateral renal vein thrombosis (RVT) will present increases, the thrombus contracts and multiple
with ARF, but is very rare. If discovered, it should collateral vessels develop.
trigger a search for a mass compressing both renal Renal veins are well demonstrated by MRI scan-
veins or the suprarenal inferior vena cava (IVC) ning, with the thrombus appearing hyperintense
and a coagulopathy screen. Unilateral RVT is more on both T1- and T2-weighted images. The kidney
frequent in contrast, but will not produce ARF if appears swollen with loss of corticomedullary dif-
both kidneys are initially normal. ferentiation on T1-weighted images, with both
Usually, a single renal vein exists on each side, cortex and medulla having low signal secondary
but up to 30% of patients will have multiple renal to prolonged T1 and T2 relaxation times.
veins. RVT is most common on the left because Venography is the gold standard and can accu-
this vein is three times longer than the right vein rately determine the presence of thrombus and its
and is more liable to extrinsic compression as it extent. A catheter inserted in the common femoral
passes between the superior mesenteric artery vein allows selection of the renal vein as it enters
and the aorta. RVT may be caused by hypercoagu- the IVC and, with contrast injection, a fi
filling defect
lable states, dehydration (particularly in infants), or complete occlusion can be demonstrated.
sepsis, or trauma and occurs in up to 40% of Therapeutic measures, such as the instillation
patients with nephrotic syndrome. of localized thrombolytics and the placement of
Ultrasound can show a distended renal vein an IVC filter if the thrombus is extensive, can be
with echogenic thrombus within that vein that performed during the diagnostic procedure.
may extend into the IVC. Absent fl flow or a thin cuff Complications include damage to vessels and
12

? ARF with previously normal kidneys or acute on chronic renal


failure
Answer with history clinical findings and Laboratory tests

Acute on chronic renal


ARF with Previously normal kidneys failure

Prerenal Renal Postrenal

Surgical
Medical
Medical Surgical
REGION
ANATOMICAL

US US

?ATN ? Aortic or renal ? Renal vein ? Glomerulonephritis or ? Obstruction


artery thrombosis acute interstitial nephritis
DDx

dissection/occlusion
CLINICAL

US – largely US US to confirm chronic


MRI or angiography to exclude changes, scars etc. +/-
if high pretest other MAG3 for function.
probability diagnoses

MRI or
Contrast enhanced Non contrast CT for
CT* obstructing mass or
MAG3 Non contrast CT for stone
? obstructing mass
TEST TO ANSWER
CLINICAL QUESTION

FIGURE 2.6. Diagnostic algorithm for imaging in ARF. US, ultrasound.


T. Sutherland
2. Imaging of Acute Renal Failure 13

dislodgement of thrombus resulting in pulmonary fi


final diagnosis to be made. Using the above dis-
embolism, and require the administration of cussion, a potential diagnostic algorithm is given
potentially nephrotoxic intravenous contrast. in Figure 2.6.
I thank Professor Alexander Pitman for his
assistance and guidance with this work.
Renal Artery Stenosis
Renal artery stenosis (RAS) is most commonly References
associated with hypertension and is usually caused 1. Liano F, Pascual J. Epidemiology of acute renal failure:
by atherosclerotic plaques and less commonly by a prospective, multicenter, community-based study.
fibromuscular dysplasia. It can exacerbate renal
fi Kidney Int 1996; 50: 811–818.
hypoperfusion and is a cause of chronic renal 2. Dahnert W. Radiology review manual. 5th ed.
impairment. In these cases, signs of chronic renal Philadelphia: Lippincott Williams & Wilkins. 2003.
impairment, as previously mentioned, will usually 3. Middleton WD, Kurtz AB, Hertzberg BS. Ultrasound—
be present. It is extremely unlikely to be the caus- The requisites. 2nd ed. St. Louis: Mosby. 2004.
ative factor for ARF presenting de novo. In patients 4. Sheafor DH et al. Non-enhanced helical CT and US
in the emergency evaluation of patients with renal
presenting with acute-on-chronic renal failure, it
colic: Prospective comparison. Radiology 2000; 217:
is one of the many possible causes of the preexist-
792–797.
ing renal failure and enters the differential diag- 5. Brant WE, Helms CA. Fundamentals of diagnostic
nosis in this fashion. radiology. 2nd ed. Philadelphia: Lippincott Williams
& Wilkins. 1999.
6. Urban BA, Ratner LE, Fishman EK. Three-dimensional
Conclusion volume-rendered CT angiography of the renal arter-
ies and veins: Normal anatomy, variants, and clinical
Clinical assessment in ARF is vital to construct a applications. Radiographics 2001; 21: 373–386.
differential diagnosis list. Imaging can then have 7. Pitman AG, Major NM, Tello R. Radiology core review.
a targeted role to answer questions and enable the Edinburgh: Saunders. 2003.
3
Drug-Induced Renal Injury
Sara Blakeley

Drug-induced nephrotoxicity contributes to 8 to 4. Monitoring of drug levels (where appropriate).


60% of all cases of in-hospital acute kidney injury 5. Discontinuation or change of mediation when
(AKI) and 1 to 23% of cases of AKI seen on the possible.
intensive care unit (ICU) (1). The wide variations 6. Consider specific
fi therapies, e.g., N-acetylcysteine
N
are caused by different definitions
fi of AKI and dif- (NAC; contrast), rasburicase, and allopurinol
ferent patient populations. Drug-induced nephrop- (tumor lysis syndrome), urinary alkalinization
athies are often underdiagnosed, and should be (drug-induced rhabdomyolysis).
considered in every type of renal failure, both acute 7. Close liaison with ICU pharmacist.
renal failure and chronic renal failure (CRF).

Nonsteroidal Anti-Inflammatory
Risk Factors for Nephrotoxicity Drugs
Many patients may be taking a potentially nephro- NSAIDs are widely used and overall renal com-
toxic drug. For example, 55% of patients admitted plications are uncommon, however, their use
to a general medical ward were taking one or in the community has been shown to increase
more of either an angiotensin-converting enzyme the risk of hospitalization with AKI by up to
inhibitor (ACEI) or angiotensin receptor antago- four-fold (4, 5). Renal effects are predominately
nist (ARB), a diuretic, or a nonsteroidal anti- caused by their effect on the production of renal
infl
flammatory drug (NSAID) (2). More than 25% of prostaglandins.
patients were taking two or three of these drugs.
Not all patients will develop renal failure; whether
a drug is nephrotoxic or not depends on patient- Effects on Renal Prostaglandins
and drug-related factors (Tables 3.1 and 3.2). Membrane-bound phospholipids are converted to
arachidonic acid by phospholipase A. Arachidonic
acid then enters the cyclooxygenase pathway
Methods of Prevention under the action of cyclooxygenase enzyme (COX)
forming prostaglandins, or enters the lipoxygen-
The following general principles should be
ase pathway, forming leukotrienes.
adhered to:
In health, renal prostaglandins have a number
1. Appropriate dose alteration in patients depending of effects: control of renin release, regulation of
on renal function, age, and ideal body weight. renal blood flow
fl (RBF) through vascular tone, and
2. Awareness of drug interactions. control of salt and water transport in the renal
3. Close attention to flfluid balance and hemody- tubules. They have both vasodilatory (prostaglan-
namic status. din [PG]-E2, PGI2) and vasoconstrictor (throm-

14
3. Drug-Induced Renal Injury 15

TABLE 3.1. Mechanism of drug-induced AKI are crucial to maintain RBF and GFR. Patients
Mechanism Drug examples who fall into this high-risk category are those with
parenchymal renal disease or renal impairment,
Altered intraglomerular Calcineurin inhibitors
hemodynamics: drugs that Vasopressors hypovolemic patients, those with congestive
interfere with the normal Amphotericin cardiac failure or liver disease (because of activa-
regulatory alterations in Contrast agents tion of the renin-angiotensin system), and the
preglomerular and postglomerular NSAIDs, ACEI/ARBs elderly. There are two isomers of the COX enzyme
arteriolar resistance can
(6). “Traditional” NSAIDs are nonselective COX
compromise renal blood flow and
glomerular filtration rate, especially inhibitors, whereas newer “coxibs” selectively
in times of hemodynamic instabilit target the production of proinfl flammatory prosta-
Drug-induced glomerulopathy: this Gold, D-penicillamine glandins by COX-2. They have fewer gastric and
usually presents with nephrotic NSAIDs platelet side effects, but it is now clear that they
syndrome or proteinuria. Renal Mesalazine
have the same effects on renal prostaglandins (7);
function need not be impaired
Drug-induced thrombotic Cyclosporin A, OKT3, therefore, caution should be still be taken in high-
microangiopathy: rare tacrolimus risk individuals.
Clopidogrel, ticlopidine NSAID-induced salt and water retention occurs
Cocaine and the effect of loop diuretics is also diminished.
Quinine
This may lead to pedal edema or hypertension, but
Direct tubular cell toxicity Aminoglycosides
Amphotericin can precipitate overt cardiac failure in at-risk
Calcineurin inhibitors patients.
Cisplatin
Methotrexate
Cocaine Other Renal Effects
Contrast media
Palmidronate 1. Acute tubulointerstitial nephritis (TIN) is
Tubular damaged caused by osmotic Mannitol less common, occurring after 3 to 5 days, or
nephrosis: tubule cells take up Dextrans even after years, of drug use. It occurs in an
nonmetabolizable molecules, Intravenous
creating an osmotic gradient. immunoglobulin
Water rapidly accumulates in Hydroxyethyl starch TABLE 3.2. Risk factors for development of drug-induced AKI
the cell, causing swelling and
vacuolation, thereby disrupting cell Patient-related factors Drug-related factors
integrity. Cellular debris causes Increased age Inherent nephrotoxic potential
tubular obstruction Preexisting chronic kidney Dose (important for drugs
Tubular damage caused by cast Statins disease inducing crystal deposition,
deposition: rhabdomyolysis Diabetes mellitus causing tubular damage,
Interstitial nephritis: drugs can cause β-lactams, quinolones, Intravascular volume depletion: and drugs altering renal
an acute allergic interstitial rifampicin, macrolides, absolute (e.g., dehydration) hemodynamics)
nephritis or chronic interstitial sulfonamides or effective (e.g., massive Prolonged duration of
damage NSAIDs ascites, nephrotic syndrome) treatment
Thiazide and loop diuretics Peripheral vascular disease Frequency of administration
Phenytoin (this increasesthe risk of Time of administration
Cimetidine, ranitidine renovascular disease)
Allopurinol Sepsis Rate of administration
Antivirals Concomitant use of diuretics (important fordrugs causing
Cocaine and other nephrotoxic drugs crystal nephropathy)
Metabolic disturbances: sodium Route of administration (e.g.,
Note: This list is not exhaustive.
depletion, hypoalbuminemia, intravenous versus oral
Source: Schetz et al., 2005; and Perazella, 2003.
acid-base disturbances (which contrast)
may exacerbate intrarenal Combination of drugs causing
crystal deposition), multiple nephrotoxic synergy (e.g.,
boxane A2) effects. In certain situations associated myeloma cephalosporins and
with high levels of circulating vasoconstrictors aminoglycosides,
aminoglycosides
vancomycin and
vancomycin,
(such as angiotensin II [ATII], endothelin, cate-
aminoglycosides)
cholamines), vasodilatory renal prostaglandins
16 S. Blakeley

idiosyncratic, non-dose-dependent manner. The ability. There is then a slow rise in serum creati-
classic features of fever, rash, arthralgia, and nine over several days.
eosinophilia are often not present. The diagnosis The risks of toxicity are increased with high
is made on renal biopsy. Treatment is to stop the initial peak serum levels, prolonged treatment,
drug and support the patient. Corticosteroids are hypovolemia, increasing age, liver dysfunction,
sometimes administered, but the evidence is hypoalbuminemia, and in combination with other
lacking. drugs (e.g., diuretics, NSAIDs, ACEIs, cephalospo-
2. Renal papillary necrosis. rins) (9).
3. Nephrotic syndrome. Once daily dosing is associated with less neph-
4. Hyperkalemia caused by hyporeninemic rotoxicity than dosing multiple times daily, but
hypoaldosteronism generally occurs in patients may not be suitable for all patient groups (e.g.,
with CRF, diabetes mellitus, and type IV renal controversial in the treatment of bacterial endo-
tubular acidosis. carditis). The risk of nephrotoxicity is reduced
when the once daily dose is given during periods
of activity (daytime) or when taking food, possi-
ACEIs and ARBs bly related to changes in urinary pH. Levels should
still be monitored carefully according to local
ATII is a potent vasoconstrictor causing constric- practice.
tion of the efferent arteriole, increasing trans-
glomerular pressure, and, thus, GFR. It also causes
systemic vasoconstriction, increasing systemic
blood pressure and improving renal perfusion. In Contrast Nephropathy
certain situations, GFR is very dependent on this
postglomerular constriction and, consequently, Incidence and Outcome
blocking this mechanism by the use of ACEI/ARBs
Contrast nephropathy (CN) is the third leading
may lead to a marked reduction in GFR (3).
cause of AKI in hospitalized patients (10), account-
If, after starting an ACEI/ARB, there is a small,
ing for 12% of cases of AKI. It is defined
fi as an
but nonprogressive rise in the serum creatinine,
acute decline in renal function, with a rise in
this is generally related to alterations in intrarenal
serum creatinine of greater than 25% from base-
hemodynamics rather than structural injury (8).
line (or an absolute rise of 44 μmol/L), occurring
If the initial serum creatinine rises by more than
24 to 48 hours after contrast. Creatinine usually
30% or if there is a progressive increase in creati-
peaks at 5 days and returns to baseline by 10 days.
nine after starting the drug, the drug should be
Incidence is varied depending on definitions
fi used
stopped and a reason sought. This situation may
and patient populations studied. Overall incidence
arise in patients with bilateral renal artery steno-
is less than 3% (11, 12), but rises in the setting
sis (or stenosis of a single functioning kidney), but
of increasing number of risk factors (13), up to
is also seen when there is a reduction in the abso-
50% with the combination of diabetes and renal
lute or effective circulating volume, in the pres-
failure (14).
ence of sepsis, and with the concomitant use of
Risk factors for development of CN:
other drugs (e.g., NSAIDs). These factors should
be addressed before restarting the drug. • Preexisting renal impairment
• Diabetes mellitus
• Absolute (e.g., dehydration) or effective volume
Aminoglycosides depletion (e.g., congestive cardiac failure,
nephrotic syndrome, liver disease)
Aminoglycoside toxicity is caused by partial • Left ventricular ejection fraction less than 40%
reabsorption of the drug by the proximal renal • Concurrent use of nephrotoxic drugs (e.g.,
tubular cells, and the first
fi indication of renal NSAIDs, aminoglycosides)
involvement is the development of polyuria • High volume, high osmolar, ionic contrast given
because of a defect in the urinary concentrating intravenously
3. Drug-Induced Renal Injury 17

• Pre procedure shock (e.g., hypotension, intra- reduce the potential for ischemic injury by inter-
aortic balloon pump) fering with active transport and decreasing the
• Increasing age oxygen demands of tubules, however, evidence is
lacking. There is no evidence that mannitol, atrial
Renal failure is normally nonoliguric and
natriuretic peptide, dopamine, or fenoldopam
resolves with supportive care. Up to 12% of
(a dopamine 1 agonist) are better than standard
patients need renal replacement therapy (11).
hydration. Calcium channel antagonists gained
The development of CN increases mortality. It
some interest but were not supported by large
is unclear whether this is because of renal failure
trials. Despite its properties as an adenosine
itself, or is simply a marker of increased disease
antagonist, there is no evidence that theophylline
severity and underlying comorbidity. In one study,
is more effective, and work continues on the anti-
the development of AKI increased mortality from
oxidant, ascorbic acid.
7 to 34% (11).

Methods of Prevention Suggestions for High-Risk Patients


Intravenous Hydration • Identify high-risk patients, correct modifiable

risks, and consider the risk/benefit
fi to the patient
Volume expansion with intravenous fluid fl before of the procedure
and after contrast is proven to be beneficial.fi • 600 to 1200 mg NAC twice daily for 2 days before
Timing, dose, and type of fluid
fl administered are procedure and two doses after the procedure
less well defined.
fi Isotonic fluid (using sodium • Intravenous hydration with saline at 1 mL/kg/h
bicarbonate) may be better than hypotonic fl fluid for 6 to 12 hours before and after the
because of better volume expansion, urinary alka- procedure
linization, and reduction of free radical-mediated
injury (15). Or
5% dextrose and H2O + 154 mEq/L sodium
N-Acetylcysteine bicarbonate at 3 mL/kg/h for 1 hour before the
procedure and 6 hours after the procedure (add
NAC counteracts renal vasoconstriction but also
154 mL of 1000 mEq/L sodium bicarbonate to
scavenges oxygen free radicals, thereby preventing
846 mL of 5% dextrose in H2O)
direct oxidative tissue damage after exposure to
• Use the minimum volume of iso-osmolar or low
contrast media (16).
osmolar contrast
Data from a series of meta-analyses are mixed
because of the lack of a standardized definition
fi of
CN, study heterogeneity, and publication bias. References
Despite this, NAC seems to reduce the risk of renal
1. Schetz M, Dasta J, Goldstein S, Golper T. Drug-
injury in high-risk patients.
induced acute kidney injury. Curr Opin Crit Care.
2005; 11: 555–565.
Type of Contrast Used 2. Loboz KK, Shenfieldfi GM. Drug combinations and
impaired renal function—the “triple whammy”.
Studies compared high, low and iso-osmolar prep-
Br J Clin Pharm. 2005; 59(2): 239–243.
arations. In high-risk patients, iso-osmolar com- 3. Perazella MA. Drug induced renal failure: Update on
pounds are associated with a reduced incidence of new medications and unique mechanisms of neph-
CN (17). Nonionic rather than ionic compounds rotoxicity. Am J Med Sci. 2003; 325(6): 349–362.
have also been found to be less nephrotoxic. 4. Perez Gutthann S, Garcia Rodriguez LA, Raiford DS,
et al. Nonsteroidal anti-infl
flammatory drugs and the
Other Therapies (18, 19) risk of hospitalization for acute renal failure. Arch
Intern Med. 1996; 156: 2433–2439.
A recent meta-analysis found no evidence that 5. Griffifin MR, Yared A, Ray WA. Nonsteroidal anti-
periprocedural renal replacement therapy reduced infl
flammatory drugs and acute renal failure in elderly
the incidence of CN (20). Loop diuretics may persons. Am J Epidemiol. 2000; 151: 488–496.
18 S. Blakeley

6. Barkin RL, Buvanendran A. Focus on the COX-1 and ing coronary angiography. Am J Med. 1990; 89(5):
COX-2 agents: Renal effects of nonsteroidal and 615–620.
anti-infl
flammatory drugs—NSAIDs. Am J Thera- 15. Merten GJ, Burgess WP, Gray LV, et al. Prevention of
peutics. 2004; 11: 124–129. contrast induced nephropathy with sodium bicar-
7. Gambaro G, Perazella MA. Adverse renal effects of bonate: a randomised controlled trial. JAMA. 2004;
anti inflflammatory agents: Evaluation of selective 291: 2328–2334.
and non selective cyclooxygenase inhibitors. J Int 16. Pannu N, Manns B, Lee H, Tonelli M. Systematic
Med. 2003; 253: 643–652. review of the impact of N-acetylcysteine on
8. Palmer BF. Renal dysfunction complicating treat- contrast nephropathy. Kidney Int. 2004; 65: 1366–
ment of hypertension. N Engl J Med. 2002; 347: 1374.
1256–1261. 17. Aspelin P, Aubry P, Fransson SG, Strasser R, Willen-
9. Beauchamp D, Labrecque G. Aminoglycoside neph- brock R, Berg KJ. Nephrotoxicity in high-risk
rotoxicity: Do time and frequency of administra- patients study of iso-osmolar and low-osmolar
tion matter? Curr Opin Crit Care. 2001; 7: 401–408. non-ionic contrast media study investigators. Neph-
10. Nash K, Hafeez A, Hou S. Hospital-acquired renal rotoxic effects in high-risk patients undergoing
insuffi
ficiency. Am J Kidney Dis. 2002; 39(5): 930–936. angiography. N Engl J Med. 2003; 348(6): 491–
11. Levy EM, Viscoli CM, Horwitz RI. The effect of acute 499.
renal failure on mortality. A cohort analysis. JAMA. 18. Maeder M, Klein M, Fehr T, Rickli H. Contrast
1996; 275(19): 1489–1494. nephropathy: Review focusing on prevention. J Am
12. Rudnick MR, Berns JS, Cohen RM, Goldfarb S. Coll Cardiol. 2004; 44: 1763–1771.
Contrast-media associated nephrotoxicity. Semin 19. Pannu N, Wiebe N, Tonelli M. Prophylaxis strategies
Nephrol. 1997; 17: 15–26. for contrast nephropathy. JAMA. 2006; 295: 2765–
13. Rich MW, Crecelius CA. Incidence, risk factors, and 2779.
clinical course of acute renal insuffi ficiency after 20. Cruz DN, Perazella MA, Bellomo R, Corradi V, de
cardiac catheterization in patients 70 years of age or Cal M, Kuang D, Ocampo C, Nalesso F, Ronco C.
older. A prospective study. Arch Intern Med. 1990; Extracorporeal blood purifi fication therapies for pre-
150(6): 1237–1242. vention of radiocontrast-induced nephropathy: a
14. Manske CL, Sprafka JM, Strony JT, Wang Y. Contrast systematic review. Am J Kidney Dis. 2006; 48(3):
nephropathy in azotemic diabetic patients undergo- 361–367.
4
Acute Kidney Injury
Sara Blakeley

Patients may be admitted to the intensive care unit three levels of renal dysfunction and two renal
(ICU) with acute kidney injury (AKI) or it may outcomes. The levels of renal dysfunction can be
develop during their stay. This chapter gives an over- defined
fi by changes in serum creatinine, GFR, or
view of the definition
fi and epidemiology of AKI, along urine output.
with clinical features and initial investigations.
Risk of Renal Dysfunction
• Serum creatinine increased 1.5 fold or
Definition of AKI • GFR decreased by more than 25% or
• Less than 0.5 mL/kg/h of urine for 6 hours
AKI is an abrupt (<7 d) and sustained decrease in Injury to the Kidney
kidney function (1). It is accompanied by changes • Serum creatinine doubled or
in blood biochemistry (e.g., a rise in serum creati- • GFR decreased greater than 50% or
nine), in urine output, or both. There is a spec- • Less than 0.5 mL/kg/h of urine for 12 hours
trum ranging from a mild transient rise in serum Failure of Kidney Function
creatinine, to overt renal failure needing renal • Serum creatinine increased 3 fold or
replacement therapy (RRT); hence, the term acute • An acute rise in creatinine of greater than
kidney injury (AKI) is more precise than the term 44 μmol/L so that new creatinine is greater
“acute renal failure”. than 350 μmol/L or
Multiple definitions
fi of ARF exist, and the reader • GFR decreased more than 75% or
is guided to a series of excellent reviews that high- • Less than 0.3 mL/kg/h of urine for 24 hours or
light this problem (1–5). A rise in serum creatinine anuria for 12 hours
is often used as a marker of renal dysfunction, but • Note: This takes into consideration acute-on-
it is affected by extrarenal factors, such as age, sex, chronic renal failure
race, and muscle bulk. It may lag behind changes Loss of Kidney Function
in glomerular filtration
fi rate (GFR), either in • Complete loss of kidney function for longer
decline or during recovery, and, therefore, does than 4 weeks
not always give a true reflection
fl of the GFR. Urine End-Stage Renal Disease
output can be used to definefi renal failure, but this • The need for dialysis for longer than 3 months
can be confounded by the use of diuretics, and
not all cases of renal failure are associated with
oliguria. Incidence and Outcome
Efforts have been made to develop a universal
and practical way of defining
fi AKI via either serum AKI develops in 5 to 7% of hospitalized patients
creatinine or urine output. One such recent pro- (6, 7). Six to 25% of patients on the ICU develop
posal is the RIFLE (5) system; an acronym for AKI (2, 8); overall, 4% of admissions require RRT.

19
20 S. Blakeley

This may underestimate the scale of the problem, nall failure) but intrarenal vasoconstriction, which
however, because when all degrees of kidney could progress to tubular damage (intrinsicc renal
dysfunction are considered using the RIFLE failure). Glomerular microthrombi are associated
criteria, 20% (9) of hospital patients and 67% of with disseminated intravascular coagulation, and
ICU patients developed some form of kidney can cause intrinsic AKI (20).
injury (10).
The incidence and progression of AKI varies
depending on the patient group studied. For Prerenal Failure
example, up to 20% of cardiac surgery patients
will develop some evidence of renal injury (11), Prerenal failure (Figure 4.1) accounts for 15 to
but only 1% will need RRT (12). 20% of cases of AKI on the ICU (13, 15). For the
AKI on the ICU is associated with a hospital kidneys to be perfused and, therefore, function,
mortality of 13 to 80% (2, 8, 10–17) and 57 to 80% adequate pressure, fl
flow, volume, and patent vessels
if RRT is needed. Renal failure rarely occurs on are needed.
its own, with up to 80% of patients with renal The kidney autoregulates to maintain a con-
failure on the ICU having another organ system stant renal blood flow (RBF) through a mean arte-
failure (8). Various factors have been associated rial pressure range of 65 to 180 mmHg. “Prerenal
with a worse outcome; including comorbidity, failure” is an appropriate, albeit exaggerated,
increased severity of illness, presence of sepsis, physiological response to renal hypoperfusion.
need for mechanical ventilation, oliguria, hospi- Stimulation of the renin-angiotensin-aldosterone
talization before ICU, and delayed occurrence of system attempts to retain salt and water and,
AKI (13–15). therefore, maintain RBF. Because renal tissue is
The development of AKI dramatically increases still preserved, once renal perfusion is restored,
mortality across all patient populations studied function should improve. A profound or prolonged
(8–10, 12, 14). Worsening levels of renal dysfunc- reduction in perfusion can, however, lead to isch-
tion, as described by the RIFLE criteria, correlate emic acute tubular necrosis (ATN) (intrinsic renal
well with increasing hospital mortality, with up to failure).
a 10-fold risk of death with “failure.” AKI carries Conditions leading to reduced renal perfusion
an independent risk of death, but it is unclear and, therefore, causing prerenal failure are:
whether this is related to the systemic effects of • Hypotension (relative or absolute) secondary to
renal failure itself, the effects of its treatment, or vasodilation (e.g., certain drugs, loss of vascular
is simply a refl
flection of the severity of the under- tone, and sepsis)
lying condition. • Compromised cardiac function
After AKI needing RRT, 10 to 32% of patients • Intravascular volume depletion (absolute or
are discharged from hospital still needing RRT effective)
(2, 16, 18). • Increased intra-abdominal pressure (abdominal
compartment syndrome)

Causes of AKI
Intrinsic Renal Failure
Causes of AKI can be divided into prerenal, intrin-
sic, and obstructive causes. One disease may be The commonest cause of intrinsic renal failure on
associated with different causes of ARF, for the ICU is ischemic ATN developing after pro-
example, sepsis is a common cause of renal dys- found or prolonged prerenal failure (Figure 4.2).
function on the ICU, accounting for up to 50% of Up to 80% of cases of AKI on the ICU are attrib-
cases of AKI. AKI occurs in 23% of patients with uted to ATN (2, 13, 15). Although ATN is a histo-
severe sepsis, and in 51% of patients with septic logical diagnosis, its development is suggested
shock when blood cultures are positive (19). Sepsis by the persistence of renal failure following the
is characterized by systemic vasodilation (prere-
( restoration of adequate renal perfusion. The
4. Acute Kidney Injury 21

Pre re
Pre
Pr rena
nall fa
na faililur
fail ure
ur e

Hypottens
Hypo tensiion
i on Hypovo
Hypo volla
laemiia
laem ia Low card
Low
Lo rdi
dia
iac out
iac outp
tputt
(Reduced intravascular volume)

Tota
Tot
Total lo
tal loss
loss Volu
Vol
Volume red
lume edi
dis
istr
ist
tribut
tributitio
ion
ion

GI los
los
osss Redu
Red
Re duce
duce
cedd ef
eff
ffe
fect
cti
tiv
ive ci
ive circ
ircullat
ati
tin
ing
ing
(Vomiti
(Vom
(V iting,
g ddiiarr
iarrhoea
iarrhhoea, surgic
i all ffiistu
istul
t lae))
vollu
vo lume
lume
(Ascit
(Asc
(A ites, oed
oedema 3rdd spacing
dema, “3 cing”
i ”, conge
congestiv
stive
ti e cardia
rdiac
di c
failure)

Haem
Haem
Ha emor
orrh
rhag
hag
ge
(Visibl
(Vis
(Vi ible and
d occult)
lt)
Altte
Al tere
tere
redd va
vasc
scul
ular
lar cap
cap
pac
acit
itan
itance
ance
( psis:
(Sep
(S i shu
h ntin
nting
ti g
g, vasod
asodil
dilattatio
ti n.
n Hepat
p torenall
synd
y drome HRS
HRS))
Rena
Rena
Re nall lo
loss
loss
(Diureti
(Di retics
tics
cs, poly
l uria
uria)
i )

Skiin
Sk in los
los
osss
(
(Exc
(E essive
essi
ive sweat
weati
ting
ing
ing,
g, bur
burns))

FIGURE 4.1. Causes of prerenal failure. GI, gastrointestinal.

Intr
Int
Intrin
tri
insi
sic
ic re
rena
nall fa
na fail
fail
ilur
lur
ure
e

Glom
Glomer
omerul
ular
lar Tubular Inte
Int
In ters
ters
rsti
titi
ti tiall Vascul
Vasc
Va ular
lar
(Glo
(Glome
lo meru
merulo
rulone
lo neph
ne phri
ph riti
ri tis)
tis) (Aut
(A utoi
ut oimm
oi mmun
mm une
un e, tox
tox
oxic
ic, in
ic infe
fect
fe ctio
ct ious
io us))
us

Ischaemic Toxic Larg


Larg
La ge ve
vess
ssell Smal
Sm allll ve
vess
ssell
(E
Ext
xtreme
xtre me pre rren
enal
al, (Ren
(Renov
ovasascu
culla
lar di
lar diseseas ase
e, ((Vas
(V
Vas
ascu
culililiti
cu tiss, HRS
ti HRS
RS,
sepsis,, pa
p nc
n re
reat
atit
ati is
itis)) athe
at hero
he roem
ro embo
em bo
olilic)
c)) reno
re nova
no vasc
vasc
scul
ular
lar ar,
r, ma
malililign
gnan
gn
nan
ant
nt
hypertension)

Intrinsic toxins
( ha
(Rhabd
bdom
bdomyo
om y lyysi
yo siss, masassi
sive
sive hae
hae
aemomoly
mo lyysi
siss
tumo
tumo
tu mour
ur llys
ysiis
ys is myel
is, elom
loma))

Extrinsic toxins
(Rad
(Radio
io
o ccon
ontr
on tras
trastt,
as t, dru
dru
rugs
gs, an
gs antititibi
biot
bi otic
ot
tic
ics)
s))

FIGURE 4.2. Causes of intrinsic renal failure (red,


d commoner causes on the ICU). HRS, hepatorenal syndrome.
22 S. Blakeley

pathophysiology of ischemic ATN is reviewed Volume Overload


elsewhere (21, 22), but an alteration in glomerular
hemodynamics with marked afferent arteriolar Salt and water retention occurs early, and is a
renal vasoconstriction causes a fall in glomerular common reason for initiating RRT on the ICU
filtration pressure and subsequently causes isch-
fi (16). Volume overload may have deleterious effects
emia. This particularly affects the outer medulla. on cardiac and respiratory function, with the
Tubular damage leads to loss of normal cell-to-cell development of peripheral edema affecting wound
adhesion and allows back leakage of filtrate
fi into healing and pressure areas.
the interstitium. Shed cells precipitate, with
protein obstructing the tubules and further com- Acidosis
promising tubular function. Local infl flammatory
mediators respond to cell injury, perpetuating the There is retention of organic anions (e.g., phos-
process. phate) and reduced production of bicarbonate
ATN can also develop secondary to a variety of by the failing tubules. In critically ill patients,
intrinsic or extrinsic renal toxins. Vascular causes this may be aggravated by the presence of a non-
of renal failure may be present at a prerenal or renal acidosis, for example, lactic acidosis from
intrarenal level, and should be considered in vas- sepsis and respiratory acidosis from respiratory
culopaths. The more classic glomerular causes for failure.
intrinsic renal failure are seen less frequently on
the ICU, but are important to recognize because
they require specificfi treatment. Electrolyte and Mineral Disturbances
Hyponatremia, hyperkalemia, and hyperphospha-
Postrenal or Obstructive Renal Failure temia are commonly seen.

Obstruction can occur at any level of the urinary


collecting system and can be caused by intrinsic Anemia
(e.g., stones, tumor) or extrinsic causes (e.g., sur-
Anemia can develop because of inappropriate
rounding or infi filtrating tumor, large infl
flammatory
levels of erythropoietin (decreased synthesis) or
abdominal aortic aneurysms). Obstruction is an
increased red cell fragility, causing premature red
infrequent cause of AKI on the ICU but is impor-
cell destruction. Uremia is also associated with
tant to be excluded in all cases.
platelet dysfunction and increased risk of gastro-
intestinal bleeding.
Complications of AKI
Immunosuppression
AKI is a systemic disease, having effects on practi-
cally all organ systems (23). It is becoming increas- Renal failure itself can impair humoral and cellu-
ingly recognized that there is “cross talk” between lar immunity, putting the patient at risk of infec-
the injured kidney and other organs through the tious complications.
release of proinfl flammatory cytokines. Complica-
tions related to other failing organs may be seen,
Metabolic Consequences
but complications specificfi to the failing kidney are
as follows. Hyperglycemia occurs because of peripheral in-
sulin resistance and increased hepatic gluconeo-
genesis. Protein catabolism is also activated.
Retention of Uremic Toxins
Accumulation of toxins, including urea, can lead
Drug Accumulation
to nausea, vomiting, drowsiness, a bleeding ten-
dency, uremic flap, and, rarely, coma (uremic Renal failure may be secondary to drugs, but, as
encephalopathy) and a pericardial rub. GFR falls, renal clearance of drugs and their
4. Acute Kidney Injury 23

metabolites also falls. Renal failure may be exac- be interpreted in light of the clinical setting, but
erbated by drug accumulation, or other side effects can act as another tool in the assessment of intra-
can develop, such as morphine metabolites leading vascular volume status.
to respiratory depression.
Radiological Investigations
A chest x-ray will help assess volume status, but
Investigation of the Cause of Renal patchy infifiltrates may also represent pulmonary
Failure (Table 4.1) hemorrhage, as seen in certain forms of vasculitis.
A renal ultrasound scan should be performed;
Laboratory Tests (Table 4.2) the timing will depend on the likely cause of renal
failure and the patient’s clinical state. Further
Urinalysis
imaging should be guided by the clinical scenario.
A standard dipstick for blood and protein should
be preformed and a fresh sample spun for casts:
hyaline casts (nonspecific
fi markers of renal injury), Conclusion
brown/cellular casts (ATN), and red cell casts
(acute glomerulonephritis). An estimation of AKI is a significant
fi condition affecting critically
protein excretion may be needed, either a 24-hour ill patients on the ICU. It is a systemic process
urine collection or a spot urine protein-creatinine affecting all organs, and has a major impact on
ratio, depending on local resources. patient morbidity and mortality. It is, therefore,
Urine osmolality and urinary electrolytes can important to be able to promptly recognize its
be used to help distinguish prerenal failure from development and institute the appropriate inves-
intrinsic kidney disease (Table 4.3). They should tigations to guide treatment.

TABLE 4.1. History and examination in AKI


A full history should be take with reference to the following:
• Presence of risk factors: known chronic renal disease, advanced age, diabetes mellitus, ischemic heart disease, hypertension, peripheral vascular
di
disease, liver
li disease,
di recentt hi
high-risk
h i k surgery
• Previous episodes of renal failure
• Family history of renal disease
• Rashes, joint aches, sinusitis, and hemoptysis suggesting a systemic condition
• Review blood pressure and anesthetic charts for periods of profound or prolonged hypotension in relation to the patients usual blood pressure
• Review fluid balance charts considering hidden losses, such as sweating or “third spacing.” The sudden onset of anuria suggests obstruction or a
catastrophic
t t hi vascular l event.t RRememberb that
th t di
diuretics
ti can make
k th
the urine
i output
t t llookk ““artificially
tifi i ll good”
d”
• Drug charts should be reviewed for intravenous contrast, chemotherapeutic agents, analgesics, antibiotics, and herbal remedies, including any
new medications taken in the past month
• Determine baseline creatine and pattern of change (i.e., sudden jump or gradual decline). The rate of change may be more important than the
absolute value
• Review any previous urinalyses for previous hematuria or proteinuria that may suggest chronic disease
A full examination should be performed with reference to the following:
• Pressure: mean arterial pressure in relation to the patients usual readings
• Flow: cardiac output studies and/or markers of end organ perfusion (e.g., lactate)
• Volume: overall volume status as well as intravascular volume status
• Patent vessels: evidence of generalized vascular disease
• Rashes or splinter hemorrhages suggesting vasculitis, cholesterol emboli, or infective endocarditis
• Palpable bladder or kidney, suggesting obstruction
• Raised intra-abdominal pressure or tense limbs, suggesting compartment syndrome
24 S. Blakeley

TABLE 4.2. Laboratory investigations for AKI


Finding Comment
Anemia Normochromic, normocytic suggests chronicity
Thrombocytopenia + microangiopathic hemolytic anemia Consider hemolytic uremic syndrome, thrombotic thrombocytopenic purpura,
disseminated intravascular coagulation (DIC)
Neutrophilia, “left shift” + thrombocytopenia Consider sepsis
Eosinophilia Consider vasculitis, allergic interstitial nephritis
Abnormal coagulation profile Sepsis, DIC, hepatorenal syndrome, systemic lupus erythematosus (SLE)
Elevated urea and creatinine Will rise with any cause of renal failure. A normal serum creatinine does not exclude
the presence of renal dysfunction, and conversely an elevated creatinine may
underestimate the degree of renal dysfunction
Elevated serum Cystatin C A newer marker of renal dysfunction, but needs further evaluation on ICU patients.
Freely filtered at the glomerulus, and fully metabolized by proximal tubular cells, if
GFR falls, levels rise
Hypercalcemia Elevated in malignancy (including hematological). Calcium may be high, low, or
normal in chronic kidney disease (CKD)
Hyperphosphatemia Extremely elevated in rhabdomyolysis and tumor lysis syndrome, but will rise with
any cause of low GFR. May be normal or high in CKD
Elevated creatinine kinase Rhabdomyolysis
Abnormal liver function tests Consider hepatorenal syndrome; sepsis; vasculitis; and hemolysis, elevated liver
enzymes, and low platelet count (HELLP) syndrome
Elevated uric acid Seen in preeclampsia but will rise with any fall in GFR
Abnormal immunoglobulins Look for myeloma and other hematological malignancies
Elevated antinuclear factor, double-stranded DNA, Investigate further for systemic diseases, such as SLE and vasculitis
antineutrophil cytoplasmic antibodies (ANCA),
antiglomerular basement membrane
antibodies, and low complement
Positive virology/serology/microbiology Certain forms of renal injury seen with specific infections, e.g., hepatitis B/C, HIV,
leptospirosis, verotoxin-producing Escherichia coli

TABLE 4.3. Urinary findings in prerenal failure and ATNa References


Prerenal Intrinsic
1. Kellum JA, Levin N, Bouman C, Lameire N. Devleop-
Urine Na <20 mmol/L >40 mmol/L
ing a concensus classification
fi for acute renal failure.
Urine : plasma (U : P) urea >20 <10
ratio
Curr Opin Crit Care. 2002; 8: 509–514.
U : P creatinine ratio >40 <10–20 2. Tillyard A, Keays R, Soni N. The diagnosis of acute
U : P osmolality >2.1 <1.2 renal failure in intensive care: mongrel or pedigree?
Specific gravity >1.020 <1.010 Anaesthesia. 2005; 60: 903–914.
Urine osmolality >500 <400 3. Hoste EA, Kellum JA. Acute kidney injury: epidemi-
Urine osmolality High (>serum + Low (<serum + ology and diagnostic criteria. Curr Opin Crit Care.
100 mOsm/L) 100 mOsm/L) 2006; 12: 531–537.
FE sodiumb <1% >1–2% 4. Mehta RL, Chertow GM. Acute renal failure defini- fi
a
These results should be interpreted with caution in patients who have had
tions and classifification: Time for a change? J Am Soc
diuretics, large volume resuscitation, the elderly, or patients with chronic Nephrol. 2003; 14: 2178–2187.
renal failure. 5. Bellomo R, Ronco C, Kellum JA et al. Acute renal
b
FE, fractional excretion of sodium: in a prerenal state, sodium is actively failure—Definition,
fi outcome measures, animal
reabsorbed by working tubules to maintain intravascular volume. The models, fluid therapy and information technology
kidney will, therefore, produce concentrated urine with a low concentra- needs. The Second International Consensus Con-
tion of sodium. Creatinine is still excreted, but relatively less sodium ference of the Acute Dialysis Quality Initiative
appears in the urine. Hence, if the tubules are intact, the amount of sodium (ADQI) group. Crit Care. 2004; 8: R204–R212.
excreted compared with creatinine (fractional excretion) falls. 6. Nash K, Hafeez A, Hou S. Hospital-acquired renal
Urine Na plasma creatinine insufficiency.
fi Am J Kid Dis. 2002; 39(5): 930–
FE Na = × 100
Plasma Na Urine creatinine 936.
4. Acute Kidney Injury 25

7. Hou SH, Bushinsky DA, Wish JB et al. Hospital- therapy on outcome in critically ill patients. Crit
acquired renal insuffi ficiency: a prospective study. Care Med. 2002; 30: 2051–2058.
Am J Med. 1983; 74(2): 243–248. 15. Brivet FG, Kleinknecht DJ, Loirat P, Landais PJ.
8. Uchino S, Kellum JA, Bellomo R et al. Beginning and Acute renal failure in intensive care units-causes,
Ending Supportive Therapy for the Kidney (BEST outcome, and prognostic factors of hospital mortal-
Kidney) Investigators. Acute renal failure in criti- ity; a prospective, multicenter study. Crit Care Med.
cally ill patients: a multinational, multicenter study. 1996; 24(2): 192–198.
JAMA. 2005; 294(7): 813–818. 16. Silvester W, Bellomo R, Cole L. Epidemiology, man-
9. Uchino S, Bellomo R, Goldsmith D et al. An assess- agement, and outcome of severe acute renal failure
ment of the RIFLE criteria for acute renal failure in of critical illness in Australia. Crit Care Med. 2001;
hospitalized patients. Crit Care Med. 2006; 34(7): 29: 1910–1915.
1913–1917. 17. Korkeila M, Ruokonen E, Takala J. Cost of care, long
10. Hoste EA, Clermont G, Kersten A et al. RIFLE crite- term prognosis and quality of life in patients requir-
ria for acute kidney injury are associated with hos- ing renal replacement therapy during intensive
pital mortality in critically ill patients: a cohort care. Int Care Med. 2000; 26: 1824–1831.
analysis. Crit Care. 2006; 10(3): R73. 18. Bagshaw SM. Epidemiology of renal recovery after
11. Kuitunen A, Vento A, Suojaranta-Ylinen R, Pettila V. acute renal failure. Curr Opin Crit Care. 2006; 12:
Acute renal failure after cardiac surgery: evaluation 544–550.
of the RIFLE classifi fication. Ann Thoracic Surg. 2006; 19. Rangel-Frausto MS, Pittet D, Costigan M et al. The
81(2): 542–546. natural history of the systemic infl flammatory
12. Chertow GM, Levy EM, Hammermeister KE et al. response syndrome (SIRS). A prospective study.
Independent association between acute renal failure JAMA. 1995; 273(2): 117–123.
and mortality following cardiac surgery. Am J Med. 20. Schrier RW, Wang W. Acute renal failure and sepsis.
1998; 104(4): 343–348. N Engl J Med. 2004; 315: 159–169.
13. Bagshaw SM, Laupland KB, Doig CJ et al. Prognosis 21. Schrier RW, Wang W, Poole B Mitra A. Acute renal
for longterm survival and renal revoery in critically failure: definitions,
fi diagnosis, pathogenesis and
ill patients with severe acute renal failure: A popu- therapy. J Clin Invest. 2004; 114: 5–14.
lation based study. Crit Care. 2005; 9: R700– 22. Lameire N, Van Biesen W, Vanholder R. Acute renal
709. failure. Lancet. 2005; 365: 417–430.
14. Metnitz PG, Krenn CG, Steltzer H et al. Effect of 23. Druml W. Acute renal failure is not a “cute” renal
acute renal failure requiring renal replacement failure! Intensive Care Med. 2004; 30: 1886–1890.
5
Medical Management of Acute Renal Failure
Nerina Harley

Acute renal failure (ARF) is both common and Patients with renal disease have a variety of
associated with signifificant mortality in the inten- clinical presentations:
sive care unit (ICU) setting (1). With an impact on
• Asymptomatic
length of stay, likelihood of survival until dis-
• Symptoms directly referable to the kidney,
charge, and cost of care, it is vital that the increas-
e.g., hematuria, flank
fl pain
ing body of evidence in critical care nephrology is
• Extrarenal symptoms, e.g., edema, hyperten-
used to refine
fi defi finitions, diagnosis, preventive
sion, uremic symptoms, consequences of
strategies, investigations, and management of
hyperkalemia
these patients (2).
• Manifestations of the underlying pathology
The medical management of ARF relies on the
or etiology, e.g., sepsis, hypotension, rhabdo-
basic tenets of diagnosis, elimination of reversi-
myolysis, systemic vasculitis
ble factors, amelioration of exacerbating factors,
treatment of complications, and optimization of
the “kidney’s environment” to provide maximal
recovery.
Investigations
Assessment of Renal Function
Diagnosis of ARF Although they lack sensitivity and specificity fi
serum creatinine and urine output are the most
Major causes of renal disease are divided, for useful parameters in clinical practice. Glomerular
simplicity, into three areas: filtration rate (GFR) assessment may be diffi
fi ficult
in nonsteady-state conditions. Comparison with
• Prerenal causes, caused by volume depletion
previous results is vital in determination of base-
with or without relative hypotension (reduced
line function, time course, and progression.
renal perfusion)
• Intrinsic renal causes, caused by vasculitis, glo-
merular disease, and tubulointerstitial disease Urinalysis
• Postrenal or obstructive causes
Urine examination may demonstrate granular and
The most common causes of ARF have been epithelial casts in ATN, eosinophils in acute inter-
found to be acute tubular necrosis (ATN) (45%), stitial nephritis, and red cell casts in acute vascu-
prerenal (21%), acute on chronic (13%), obstruc- litis or glomerulonephritis and/or proteinuria.
tion (10%), glomerulonephritis (GN), or vasculitis There are no prospective studies of the predictive
(4%) (3). value of urine sediment in ATN. Urinary elec-
Risk factors for the development of ARF are trolytes are of limited value in most clinical
given in Table 5.1. situations because of the confounding effects of

26
5. Medical Management of Acute Renal Failure 27

TABLE 5.1. Risk factors for ARF inherent risks of bleeding should be weighed up
Risk factor Selected reference(s) in the setting of the risk-to-benefit
fi ratio.
Note: The “gold standard” for the diagnosis of
Older age Nash et al., 2002 (43)
Diabetes Parfrey et al., 1989 (44). a prerenal cause of ARF is resolution of renal
Underlying renal insufficiency impairment in response to fluid challenge. This is
Cardiac failure in contrast to significant
fi ATN, in which there is
Sepsis Brun-Buisson et al., 2004 (45) prolonged time to resolution.
Ricci et al., (46)
Absolute or relative hypovolemia
Hepatic failure Han and Hyzy, 2006 (47)
Nephrotoxins (including Barrett et al., 1993 (48) Primary Prevention
high-osmolality Aspelin et al., 2003 (49).
radiocontrast agents) McCullogh et al., 2006 (50) Primary prevention of ARF in the critically ill with
Cardiopulmonary bypass with Mangano et al., 1998 (51)
or without baseline risk factors consists of avoid-
aortic cross-clamping Chertow et al., 1998 (52)
(particularly valve surgery) ance, amelioration, and treatment of these factors
Nonrenal organ transplantation Lima et al., 2003 (53) wherever possible. Strategies can be divided into
Abdominal compartment syndrome McNelis et al., 2003 (54) nonpharmacological, for example, fluid
fl adminis-
tration to reduce the risk of contrast induced
nephropathy (6), and pharmacological. To date, no
treatments such as diuretics. Urine volume is of pharmacological strategies have conclusively dem-
little diagnostic value, although little or no output onstrated prevention of ARF from any insult (7).
is acutely useful with causes of anuria, including
shock, bilateral urinary obstruction, renal cortical
necrosis, and bilateral vascular occlusion.
Loop Diuretics
Although oliguria (<400 mL/24 h) is common in
ATN, anuria is rare and other etiologies must be
Other Markers of Renal Injury
considered. Nonoliguric patients have a better
These have been described but are not currently prognosis than oliguric patients in terms of a
generally available in the acute setting. For greater residual GFR (8), lower peak serum creati-
example, serum cystatin C has been recently nine, and dialysis-free survival at 21 days (9, 10),
proposed as a marker of ARF (4), predicting ARF possibly reflecting
fl less severe renal injury. A clini-
by at least 24 hours. cal issue of the use of loop diuretics often arises in
increasing the urine output of oliguric patients.
Experimentally, loop diuretics reduce active
Radiology
sodium chloride transport in the thick ascending
Renal ultrasound is the modality of choice to limb of the loop of Henle, decreasing energy
exclude obstruction. Reduced renal size and corti- requirements and, thus, protecting the cell in
cal thinning (although preserved in diabetic the setting of decreased energy availability. Only
nephropathy) is indicative of chronic renal impair- anecdotal human evidence suggests that the use
ment. A helical computed tomographic (CT) scan of loop diuretics may be beneficialfi in the first 24
may be useful in urolithiasis, but there are risks of hours in flushing tubular casts. There is no evi-
secondary injury with radiocontrast. dence of benefi fit in established ATN on duration
of renal failure, requirement for dialysis, or sur-
vival (11). The increase in urine output in this
Renal Biopsy
setting is caused by decreased tubular reabsorp-
Renal biopsy is considered when noninvasive eval- tion in residual functioning nephrons (not re-
uation has not established the diagnosis (5); the cruitment of nonfunctioning nephrons), volume
major indications include isolated hematuria with expansion (initial sodium retention), and urea
proteinuria, nephrotic syndrome, acute nephritic osmotic diuresis.
syndrome, and unexplained ARF. Percutaneous A number of studies have suggested worsened
biopsy is most commonly performed and the outcomes in ATN with the use of loop diuretics in
28 N. Harley

the setting of contrast media (9, 12) and cardiac The incidence of ARF and requirement for
surgery. A systematic review (13, 14) comparing RRT was not significantfi different between
fluids alone with diuretics found no evidence of
fl groups (20).
improvement in survival, incidence of ARF, or • A large randomized controlled trial of low-dose
need for renal replacement therapy (RRT). Deaf- dopamine in 328 critically ill patients with
ness, possibly permanent, is a known complica- impaired creatinine, oliguria, and at least two
tion of high-dose loop diuretics. systemic infl flammatory response syndrome
(SIRS) criteria failed to show any benefit fi in
progression, need for RRT, or death (21).
Mannitol • Studies of fenoldopam, a relatively selective
Mannitol may preserve mitochondrial function by dopamine A1 receptor agonist, although shown
minimizing postinjury edema. Human trials have to increase sodium excretion and renal blood
failed to show benefifit in reducing ARF with man- flow in healthy and hypertensive patients, has

nitol versus fluid alone in rhabdomyolysis (15), not shown benefi fit in ARF in the critically ill
cardiac surgery (16), and vascular and biliary tract (22).
surgery (17). A trend toward harm was noted in
the prevention of contrast nephropathy (12).
N-Acetylcysteine
N-Acetylcysteine (NAC) has been shown in a
N
Dopamine Agonists number of studies to decrease the incidence of
Dopamine has a number of effects in the kidney contrast nephropathy in high-risk patients (23,
via dopamine A1 and A2 receptors. In the proxi- 24), but without improvement in RRT require-
mal tubule, dopamine, via the generation of cyclic ment or survival. Importantly, NAC may decrease
AMP, decreases Na+-H+ exchange and the Na+-H+- creatinine via activation of creatinine kinase (25)
ATPase pump, thus, decreasing sodium reabsorp- but not GFR. Promising studies have led to the
tion. In the collecting tubules, this effect on introduction of protocols in many institutions for
Na+-H+-ATPase and decreased aldosterone secre- the prophylactic use of NAC in the prevention
tion reduces sodium reabsorption (18). of contrast-induced nephropathy. With few side
When infused in doses of 0.5 to 3 μg/kg/min, effects, low cost, and ease of administration (oral
dopamine causes afferent and efferent glomerular or intravenous), many centers have erred on the
arteriolar dilatation, increasing blood flow
fl with side of possible benefit
fi in the face of lack of hard
little or no increase in GFR. At higher concentra- evidence of long-term benefit fi (26).
tions, dopamine causes vasoconstriction via α-
adrenergic receptors.
There is no evidence in human studies for a Others
“renal protective effect” of dopamine.
Trials have failed to demonstrate benefi fit of natri-
• In 1994, Baldwin et al. studied the effect of post- uretic peptides (10, 27) or adenosine agonists (28).
operative low-dose dopamine on renal function Experimental therapies, such as antioxidants and
after major elective vascular surgery. Patients erythropoietin are unproven in humans.
were administered saline or saline plus dopa- In the absence of further evidence, flfluids, and
mine as fluid replacement. No difference in renal possibly NAC, are the gold standards of interven-
function was demonstrated between the two tional strategies.
groups (19).
• In the North American Study of the Safety
and Efficacy
fi of Murine Monoclonal Antibody Supportive Strategies
to Tumor Necrosis Factor for the Treatment
of Septic Shock (NORASEPT) II study, 400 Fluid resuscitation and correction of hypotension
patients with septic shock and oliguria nonran- are clearly essential. There is no evidence of
domly received no, low-, or high-dose dopamine. advantage of one particular inotrope over another.
5. Medical Management of Acute Renal Failure 29

No evidence exists that reversing hypotension Assessment and Correction of


with noradrenaline compromises mesenteric or Volume Depletion
renal blood flow (29).
Van den Berghe demonstrated that tight glucose Clinical signs of relative or absolute volume deple-
control with intravenous therapy improved out- tion include loss of tissue turgor, hypotension,
comes in critically ill patients, including decreased postural hypotension, and decreased venous pres-
incidence of ARF (30, 31). sure (reduced jugular venous pressure). Although
Nutritional supportt of the critically ill is the left ventricular end diastolic pressure (LVEDP)
basic standard of care, although not always is the most important determinant of left ventricu-
achieved. Early enteral nutrition is supported by lar output and, thus, tissue perfusion, central
meta-analyses of Level II trials; benefits fi include venous pressure is useful because it has a direct
preservation of muscle mass, the maintenance of relationship to LVEDP, with the exceptions of
the gastrointestinal mucosal barrier and immune pure left-sided or pure right-sided failure (cor
status, and a possible reduction in multiorgan pulmonale). Other clinical manifestations may
dysfunction (32–34). A Level I trial is currently be specifi fic to the source of depletion (losses
planned by the Australian and New Zealand Inten- or third space sequestration), type of fl fluid lost,
sive Care Society (ANZICS) clinical trials group and to the associated electrolyte and acid-base
comparing early enteral nutrition with standard abnormalities.
care in 1470 critically ill patients intolerant of Recently, Vincent proposed a protocol for
early enteral nutrition (www.actr.org.au). routine flfluid challenge with defi fined rules based
There is no evidence to support prophylactic on clinical response to the volumes infused,
hemofiltration
fi to prevent contrast nephropathy, allowing for prompt deficit fi correction while
despite fifiltration removal of contrast. minimizing risks of fluid
fl overload (38).
Early recognition and adequate management of
the deteriorating clinical condition of a patient is
fundamental in the prevention of morbidity and Maintenance of Adequate MAP and
mortality, including ARF. In a hospital-based Cardiac Output
study of a medical emergency team (MET) system,
Bellomo et al. demonstrated reduced incidence of There is currently insufficient
fi data to recommend
postoperative adverse outcomes, mortality rate, firm therapeutic targets, suffi
fi fice to say, Level III
and mean hospital length of stay (35). This was evidence suggests failure to maintain systolic
not validated in a larger cluster-randomized con- blood pressure greater than 80 mmHg or MAP
trolled trial of 23 hospitals with no effect on inci- greater than 50 mmHg is associated with increased
dence of cardiac arrest, unplanned admissions to risk of developing ARF (39). A low cardiac output
ICU, or unexpected death (36). Nevertheless, the is a major risk factor for ARF after cardiac surgery
principles of early recognition, monitoring, and (40), but supranormal cardiac output has no
adequate response remain fundamental. beneficial
fi effect.

Postinjury Prevention of ARF Avoidance of Further Insults


Appropriate dosage of medication and avoidance
Secondary renal injury occurs after the primary (or protective strategies) of nephrotoxins is
insult has triggered the initial injury to the advised.
kidneys.
Strategies for postinjury prevention of ARF
overlap with those of primary prevention (37) as
Renal Replacement Therapy
described above. They include maintenance of
adequate intravascular volume, cardiac output, Intermittent versus continuous renal replacement
mean arterial blood pressure (MAP), avoidance of strategies and dose delivery are discussed in a
further insult, and supportive strategies. later chapter.
30 N. Harley

TABLE 5.2. Summary of guidelines for management of ARF


Make diagnosis
Exclusion of prerenal causes E.g., volume depletion, cardiac and liver disease, nephrotoxins
Exclusion of postrenal causes Renal ultrasound
Exclusion of intrinsic renal causes E.g., review urinary sediment, consider renal biopsy
Evaluation of urinary electrolytes Only in the absence of diuretics

Treat reversible causes


Volume resuscitation Maintain fluid balance but avoid overload
Blood pressure support Inotropic if necessary, however no validated physiological targets or end points have been established
Treat electrolyte complications E.g., hyperkalemia
No dopamine

Address/avoid exacerbating factors


Treatment of underlying etiology E.g., treat sepsis, further investigation if diagnosis unclear, surgical referral if appropriate
Adjust medication dosage Discontinue any nephrotoxic drugs
Avoid further renal insult E.g., minimize contrast-induced injury with fluids and consider NAC

Optimize “kidney’s environment”


Maintain renal perfusion Maintain fluid balance, blood pressure, and cardiac output
Adequate nutrition
Glucose control
Appropriate RRT Timely introduction of RRT, avoidance of complications (e.g., hypotension, line-related sepsis,
biocompatible dialysis membranes), and appropriate dose of dialysis

Other
Constantly review diagnosis, which may Consider further investigations, e.g., renal biopsy and radiological investigations, as appropriate
multifactorial in nature
Appropriate medical review E.g., MET resources, nephrology input

Summary 3. Liano G, Pascual J. Acute renal failure. Madrid Acute


Renal Failure Study Group. Lancet. 1996; 347(8999):
479.
Measures of severity of illness are limited when
4. Herget-Rosenthal S, Marggraf G, Husing J, et al.
applied to patients with ARF and, at present, none Early detection of acute renal failure by serum cys-
are currently adequate to predict mortality of ARF tatin C. Kidney Int. 2004; 66(3): 1115–1122.
(41). In the light of available evidence, guidelines 5. Madaio MP. Renal biopsy. Kidney Int. 1990; 38(3):
for the management of ARF have been formulated 529–543.
and given in Table 5.2. Admiral efforts to clearly 6. Mueller C, Buerkle G, Buettner HJ, et al. Prevention
defi
fine levels of injury in ARF and physiological of contrast media-associated nephropathy: ran-
end points (42) will enable further research into domized comparison of 2 hydration regimens in
prevention, amelioration, and management of 1620 patients undergoing coronary angioplasty.
ARF and, thus, impact on the significant
fi associ- Arch Intern Med. 2002; 162(3): 329–336.
ated morbidity and mortality. 7. Kellum JA, Leblanc M, Gibney RT, et al. Primary
prevention of acute renal failure in the critically ill.
References Curr Opin Crit Care. 2005; 11(6): 537–541.
8. Rahman SN, Conger JD. Glomerular and tubular
1. Levy EM, Viscoli CM, Horwitz RI. The effect of acute factors in urine flow rates of acute renal failure
renal failure on mortality. A cohort analysis. JAMA. patients. Am J Kidney Dis. 1994; 23(6): 788–793.
1996; 275(19): 1489–1494. 9. Lassnigg A, Donner E, Grubhofer G, et al. Lack of
2. Kellum JA. Acute renal failure, interdisciplinary renoprotective effects of dopamine and furosemide
knowledge and the need for standardization. Curr during cardiac surgery. J Am Soc Nephrol. 2000;
Opin Crit Care. 2005; 11(6): 525–526. 11(1): 97–104.
5. Medical Management of Acute Renal Failure 31

10. Allgren RL, Marbury TC, Rahman SN, et al. Anarit- 23. Birck R, Krzossok S, Markowetz F, et al. Acetylcys-
ide in acute tubular necrosis. Auriculin Anaritide teine for prevention of contrast nephropathy: meta-
Acute Renal Failure Study Group. N Engl J Med. analysis. Lancet. 2003; 362(9384): 598–603.
1997; 336(12): 828–834. 24. Pannu N, Manns B, Lee H, Tonelli M. Systematic
11. Cantarovich F, Rangoonwala B, Lorenz H, et al. review of the impact of N-acetylcysteine on contrast
High-dose furosemide for established ARF: a nephropathy. Kidney Int. 2004; 65(4): 1366–1374.
prospective, randomized, double-blind, placebo- 25. Genet S, Kale RK, Baquer NZ. Effects of free radicals
controlled, multicenter trial. Am J Kidney Dis. 2004; on cytosolic creatine kinase activities and protec-
44(3): 402–409. tion by antioxidant enzymes and sulfhydryl com-
12. Solomon R, Werner C, Mann D, et al. Effects of pounds. Mol Cell Biochem. 2000; 210(1–2): 23–28.
saline, mannitol, and furosemide to prevent acute 26. Bagshaw SM, McAlister FA, Manns BJ, Ghali WA.
decreases in renal function induced by radiocon- Acetyl-cysteine used in the prevention of contrast-
trast agents. N Engl J Med. 1994; 331(21): 1416– induced nephropathy: a case study in the pitfalls in
1420. the evolution of evidence. Arch Intern Med. 2006 Jan
13. Kellum JA. Diuretics in acute renal failure: pro- 23; 166(2): 161–166.
tective or deleterious. Blood Purif. f 1997; 15(4–6): 27. Lewis J, Salem MM, Chertow GM, et al. Atrial natri-
319–322. uretic factor in oliguric acute renal failure. Anarit-
14. Kellum JA. The use of diuretics and dopamine in ide Acute Renal Failure Study Group. Am J Kidney
acute renal failure: a systematic review of the evi- Dis. 2000; 36(4): 767–774.
dence. Crit Care (Lond). 1997; 1(2): 53–59. 28. Kramer BK, Preuner J, Ebenburger A, et al. Lack of
15. Homsi E, Barreiro MF, Orlando JM, Higa EM. renoprotective effect of theophylline during aorto-
Prophylaxis of acute renal failure in patients coronary bypass surgery. Nephrol Dial Transplant.
with rhabdomyolysis. Ren Fail. 1997; 19(2): 283– 2002; 17(5): 910–915.
288. 29. Levy B, Bollaert PE, Charpentier C, et al. Compari-
16. Ip-Yam PC, Murphy S, Baines M, et al. Renal func- son of norepinephrine and dobutamine to epineph-
tion and proteinuria after cardiopulmonary bypass: rine for hemodynamics, lactate metabolism, and
the effects of temperature and mannitol. Anesth gastric tonometric variables in septic shock: a pro-
Analg. 1994; 78(5): 842–847. spective, randomized study. Intensive Care Med.
17. Gubern JM, Sancho JJ, Simo J, Sitges-Serra A. A ran- 1997; 23(3): 282–287.
domized trial on the effect of mannitol on postop- 30. Van den Berghe G, Wouters P, Weekers F, et al. Inten-
erative renal function in patients with obstructive sive insulin therapy in the critically ill patients.
jaundice. Surgery. 1988; 103(1): 39–44. N Engl J Med. 2001; 345(19): 1359.
18. Denton MD, Chertow GM, Brady HR. “Renal-dose” 31. Van den Berghe G, Wouters PJ, Bouillon R, et al.
dopamine for the treatment of acute renal failure: Outcome benefi fit of intensive insulin therapy in the
scientifific rationale, experimental studies and clini- critically ill: Insulin dose versus glycemic control.
cal trials. Kidney Int. 1996; 50(1): 4–14. Crit Care Med. 2003; 31(2): 359–366.
19. Baldwin L, Henderson A, Hickman P. Effect of post- 32. Doig GS, Simpson F. Evidence-based guidelines for
operative low-dose dopamine on renal function nutritional support of the critically ill: results of
after elective major vascular surgery. Ann Intern a bi-national guideline development conference.
Med. 1994; 120(9): 744–747. Sydney: EvidenceBased.net; 2005. Download www.
20. Marik PE, Iglesias J. Low-dose dopamine does not EvidenceBased.net.
prevent acute renal failure in patients with septic 33. Dhaliwal R, Heyland DK. Nutrition and infection in
shock and oliguria. NORASEPT II Study Investiga- the intensive care unit: what does the evidence show?
tors. Am J Med. 1999; 107(4): 387–390. Curr Opin Crit Care. 2005; 11(5): 461–467.
21. Bellomo R, Chapman M, Finfer S, et al. Low-dose 34. Doig GS, Simpson F. Early enteral nutrition in the
dopamine in patients with early renal dysfunction: critically ill: do we need more evidence or better
a placebo-controlled randomised trial. Australian evidence? Curr Opin Crit Care. 2006; 12(2): 126–
and New Zealand Intensive Care Society (ANZICS) 130.
Clinical Trials Group. Lancet. 2000; 356(9248): 35. Bellomo R, Goldsmith D, Uchino S, Buckmaster J,
2139–2143. Hart G, Opdam H, Silvester W, Doolan L, Gutteridge
22. Tumlin JA, Finkel KW, Murray PT, et al. Fenoldopam G. Prospective controlled trial on the effect of
mesylate in early acute tubular necrosis: a random- medical emergency team on postoperative morbid-
ized, double-blind, placebo-controlled clinical trial. ity and mortality rates. Crit Care Med. 2004; 32(4):
Am J Kidney Dis. 2005; 46(1): 26–34. 916–921.
32 N. Harley

36. Hillman K, Chen J, Cretikos M, Bellomo R, Brown 45. Brun-Buisson C, et al. EPISEPSIS: a reappraisal of
D, Doig G, Finfer S, Flabouris A; MERIT study inves- the epidemiology and outcome of severe sepsis in
tigators. Introduction of the medical emergency French intensive care units. Intensive Care Med.
team (MET) system: a cluster-randomised con- 2004; 30(4): 580–588.
trolled trial. Lancet. 2005; 365 (9477): 2091–2097. 46. Ricci Z, et al. Practice patterns in the management
37. Bellomo R, Bonventre J, Macias W, Pinsky M. Man- of acute renal failure in the critically ill patient: an
agement of early acute renal failure: focus on post- international survey. Nephrol Dial Transplant. 2006;
injury prevention. Curr Opin Crit Care. 2005; 11(6): 21(3): 690–696.
542–547. 47. Han MK, Hyzy R. Advances in critical care manage-
38. Vincent JL, Weil MH. Fluid challenge revisited. Crit ment of hepatic failure and insufficiency.
fi Crit Care
Care Med. 2006; 34(5): 1333–1337. Med. 2006; 34(9 Suppl): S225–231.
39. Kohli HS, Bhaskaran MC, Muthukumar T, et al. 48. Barrett B, et al. Meta analysis of the relative nephro-
Treatment-related acute renal failure in the elderly: toxicity of high- and low-osmolality iodinated con-
a hospital-based prospective study. Nephrol Dial trast media. Radiology. 1993; 188(1): 171–178.
Transplant. 2000; 15(2): 212–217. 49. Aspelin P, et al. Nephrotoxic effects in high-risk
40. Suen WS, Mok CK, Chiu SW, et al. Risk factors for patients undergoing angiography. N Engl J Med.
development of acute renal failure (ARF) requiring 2003; 348(6): 491–499.
dialysis in patients undergoing cardiac surgery. 50. McCullogh PA, et al. Risk prediction of contrast-
Angiology. 1998; 49(10): 789–800. induced nephropathy. Am J Cardiol. 2006; 98(68):
41. Uchino S, Bellomo R, Morimatsu H, et al. External 27K–36K.
validation of severity scoring systems for acute 51. Mangano C, et al. Renal dysfunction after myocar-
renal failure using a multinational database. Crit dial revascularization: risk factors, adverse out-
Care Med. 2005; 33(9): 1961–1967. comes, and hospital resource utilization. Ann Intern
42. Murray PT, Le Gall JR, Dos Reis Miranda D, et al. Med. 1998; 128(3): 194–203.
Physiologic endpoints (effi ficacy) for acute renal 52. Chertow G, et al. Independent association between
failure studies. Curr Opin Crit Care. 2002; 8(6): acute renal failure and mortality following cardiac
519–525. surgery. Am J Med. 1998; 104(4): 343–348.
43. Nash K, et al. Hospital-acquired renal insufficiency.
fi 53. Lima E, et al. Risk factors for development of acute
Am J Kidney Dis. 2002; 39(5): 930–936. renal failure after liver transplantation. Ren Fail.
44. Parfrey P, et al. Contrast material-induced renal 2003; 25(4): 553–560.
failure in patients with diabetes mellitus, renal 54. McNelis J, et al. Abdominal compartment syndrome:
insuffificiency, or both. A prospective controlled clinical manifestations and predictive factors. Curr
study. N Engl J Med. 1989; 320(3): 143–149. Opin Crit Care. 2003; 9(2): 133–136.
6
Acute Renal Failure in the Surgical Patient
Marlies Ostermann

Acute renal failure (ARF) is a potential complica- outer medulla is metabolically very active, despite
tion of any surgical procedure. In general, the risk a relatively low oxygen delivery. As a consequence,
of ARF is increased in patients with underlying oxygen extraction in this region is approximately
vascular disease, diabetes mellitus, or chronic 80% and, in the event of ischemia, this area is often
kidney disease. High-risk situations include car- the first part of the kidney to suffer injury.
diovascular, hepatobiliary, and trauma surgery,
especially if performed as an emergency.
Risk Factors for ARF in the Surgical
Pathophysiology of ARF Patient (Table 6.1)

The most common causes of postoperative renal


Effect of Anesthesia
failure are hypotension (relative and absolute) Most inhalation and intravenous anesthetics cause
and/or volume depletion. Renal oxygen consump- some degree of cardiac depression and/or vasodi-
tion is determined by blood flow, making the lation, resulting in lowering of the blood pressure.
kidneys particularly vulnerable to ischemic injury Similarly, spinal or epidural anesthesia can cause
when flow is reduced. The kidneys receive 20 to hypotension as a result of increased venous ca-
25% of the cardiac output. In healthy individuals, pacitance and arterial vasodilation. In response,
renal blood flow
fl is regulated by a complex inter- compensatory changes lead to renal arterial vaso-
play between intrinsic autoregulation, and hor- constriction and increased retention of sodium
monal and neuronal influences.
fl This results in a and water. Maintenance of an adequate intravas-
relatively constant renal blood fl flow when mean cular volume and blood pressure in the range of
arterial blood pressure (MAP) is 80 to 180 mmHg. patient’s baseline blood pressure is essential to
Outside of these limits, renal blood fl
flow becomes prevent anesthesia-related renal impairment.
pressure dependent, and glomerular filtration

ceases when MAP is less than 40 to 50 mmHg. In
patients with long-standing hypertension, this
Effect of Drugs
mechanism of autoregulation is lost, and renal Certain medication may be directly nephrotoxic
function is pressure dependent, often needing a (e.g., aminoglycosides, amphotericin, nonsteroi-
MAP of greater than 80 mmHg. As a result, these dal anti-infl flammatory drugs) or may alter intra-
patients need higher blood pressures to maintain renal hemodynamics (e.g., angiotensin-converting
glomerular filtration
fi and are particularly vulner- enzyme inhibitors, nonsteroidal anti-inflamma-fl
able to hypotension or volume depletion. tory drugs). In rare situations, a drug-induced
The whole kidney only extracts less than 10% interstitial nephritis may occur. This is most com-
of the oxygen carried through the kidney. The monly caused by antibiotics or nonsteroidal anal-
thick ascending limb of the loop of Henle in the gesics, but any drug can cause interstitial nephritis.

33
34 M. Ostermann

TABLE 6.1. Risk factors for ARF developing in surgical patients 25% or a fall in glomerular filtration rate by 25%.
Preoperative factors One to 5% of patients need renal replacement
State of hydration/adequate resuscitation preoperatively therapy. A combination of patient-specific fic comor-
Contrast media bidities and factors related to surgery usually con-
Preexisting sepsis tribute to the development of ARF.
Patient comorbidity
Patient-specific
fic risk factors may not be imme-
• Advanced age
• Diabetes mellitus diately obvious. A large study on 7310 patients
• Hypertension undergoing coronary artery bypass grafting
• Cardiovascular disease (CABG) demonstrated that 29.6% of patients
• Preexisting renal impairment reported being diabetic, but an additional 5.3% of
Operative factors patients were found to have previously undiag-
Related to anesthesia (see text) nosed diabetes. Similarly, among patients under-
Related to surgery going coronary angiography, 12% of patients were
• Emergency surgery
• Duration of cardiopulmonary bypass
found to have an undiagnosed renal artery steno-
• Clamping of renal arteries sis of greater than 75%.
• Suprarenal aortic cross clamping Risk factors related to surgery tend to be
Postoperative factors less predictable, but the risk of renal failure is
• Sepsis generally higher in patients undergoing combined
• Bleeding CABG and valve replacement compared with
• Nephrotoxic drugs patients who only need one procedure.
• Contrast media
Very recently, aprotinin was identified
fi as a risk
• Cardiovascular complications associated with a fall in cardiac
output
t t ((e.g., myocardial
di l iinfarction,
f ti pulmonary
l embolus,
b l factor for ARF. An observational study on 4374
pericardial effusion) patients undergoing revascularization showed
• Development of intra-abdominal compartment syndrome that the use of aprotinin was associated with a
doubling of the risk of renal failure requiring
dialysis when compared with aminocaprionic
Treatment consists of discontinuation of the acid, tranexamic acid, or no antifibrinolytic.

offending drug and possibly steroid therapy.

Methods of Prevention of ARF


Development of Intra-abdominal
Compartment Syndrome Several studies have focussed on prevention of
ARF after cardiac surgery. To date, no magic bullet
Patients with severe ileus, bowel obstruction, has been identifi fied. Prophylactic therapy with
pancreatitis, or after trauma can develop increased dopamine, mannitol, theophylline, and diuretics
abdominal pressure leading to increased pressure has not been effective. There is some evidence that
in renal veins and renal parenchyma resulting in diuretic use after cardiac surgery increases the
decreased renal perfusion. There is a strong cor- risk of ARF. Data regarding the benefits fi of off-
relation between intra-abdominal pressure and pump surgery in patients at high risk of renal
ARF, with oliguria developing when the intra-
abdominal pressure is greater than 15 mmHg and
anuria developing when pressure is greater than TABLE 6.2. Risk factors after cardiac surgery
30 mmHg. Intra-abdominal decompression is the
Patient specific Surgery related
treatment of choice.
• Advanced age • Emergency surgery
• Diabetes mellitus • Reoperation
Type of Surgery • Hypertension • Prolonged duration of
• Preexisting renal impairment cardiopulmonary bypass
• Impaired left ventricular function • Reexploration for bleeding
Cardiac Surgery (Table 6.2) • Pericardial tamponade
• Deep sternal or systemic
Twenty to 40% of patients undergoing cardiac
infection
surgery experience a rise in serum creatinine by
6. Acute Renal Failure in the Surgical Patient 35

TABLE 6.3. Specific tests to consider in determining the cause of ARF in surgical patientsa
Blood tests Creatinine kinase To exclude rhabdomyolysis especially after trauma
Full blood count Eosinophilia is seen in 80% of patients with drug-induced interstitial
nephritis
Urinalysis Dipstick Significant proteinuria/hematuria/casts suggest intrinsic renal
pathology
Biochemistry Urinary sodium and osmolality to help differentiate prerenal failure
from ATN
Culture To exclude urinary tract infection
Diagnostic imaging Ultrasound scan To exclude obstruction and to determine renal size
Imaging of renal perfusion (e.g., renal Renal vascular supply may be of concern after major abdominal aortic
Dopplers, computed tomographic surgery. Investigation will depend on patient stability and local
angiogram, MAG3, DTPA) resources
Measurement of intravesical To exclude intra-abdominal hypertension and compartment syndrome
pressure
a
MAG3, mercaptoacetyltriglycine; DTPA, diethylene triamine pentaacetic acid.

failure is still confl


flicting. Although low cardiac volume depletion, bleeding complications, and/or
output has been shown to be a strong risk factor urosepsis.
for ARF after cardiac surgery, there is no evidence
that increasing cardiac output from adequate to
supranormal has beneficial
fi renal effects. Diagnosis (Table 6.3)

Vascular Surgery At present, there are no universally accepted cri-


teria for the definition
fi of ARF. Most arbitrary defi fi-
Patients undergoing vascular surgery generally nitions are based on a rise in serum creatinine or
represent a high-risk group for renal failure. The fall in calculated creatinine clearance. It is impor-
exact incidence of renal injury in this context is tant to remember that the glomerular fi filtration
unknown but, again, depends on the definition
fi of rate has to fall to less than 50% before serum
renal injury, patient characteristics, and type of creatinine rises, which means that any rise in
surgery. Open surgical repair of abdominal aneu- serum creatinine always implies significant
fi renal
rysms is associated with a high risk of renal failure, injury.
especially with suprarenal aortic cross clamping,
massive bleeding, or cholesterol embolization.
The emergence of endovascular repair has led to
a reduced incidence of ARF, however, the risk is
Treatment
not completely abolished by vascular stents. In
General Measures
rare instances, endovascular stents have been
found to migrate, resulting in occlusion of arterial • Restoration of renal perfusion pressure
orifi
fices, including renal arteries. • Correction of volume depletion
• Avoidance of hypotension (including relative
hypotension). Be guided by the patient’s preex-
Urological Surgery isting blood pressure
Obstruction is a common cause of ARF in patients • Fluid resuscitation, as appropriate, and use of
with urological problems. Although this diagnosis vasopressor agents if perfusing pressure still
is usually made before surgery, it may occur inadequate
postoperatively (e.g., after renal transplantation • Optimal treatment of sepsis/septic shock
or ureteric surgery). In general, the majority of • Avoidance of nephrotoxic medication if possi-
ARF posturological surgery is because of acute ble. Close monitoring of drug levels when ami-
tubular necrosis precipitated by hypotension, noglycosides are necessary
36 M. Ostermann

• Renal replacement therapy in case of severe Early Resuscitation


metabolic acidosis, unresponsive fluid
fl overload,
resistant hyperkalemia, or pericarditis Early recognition of patients at risk and timely
• Early involvement of nephrologists if an intrinsic resuscitation has been shown to result in signifi-

cause of renal failure is a possibility or if the cant reduction of the development of ARF.
patient is likely to require ongoing renal support
Prevention of Contrast-Induced
Specific Measures Nephropathy
• Removal of septic focus if possible (e.g. drainage The administration of fluids has been shown to be
of intra-abdominal abscess) the most important factor in prevention of con-
• Relief of obstruction trast-induced renal injury. Although the optimal
• Management of abdominal compartment syn- fluid regimen is uncertain, available data support

drome (including consideration of abdominal a regimen of 0.9% saline at 1 mL/kg/h intrave-
decompression) nously from up to 12 hours before administration
• Revascularization of kidneys, if appropriate of contrast medium and for up to 12 hours after.
Studies on the prophylactic role of N-acetylcyste-
N
Treatments that Have Not Been Shown to ine have had confl flicting results. Meta-analyses
Alter the Course of ARF have concluded that prophylactic N-acetylcysteine
N
was harmless and may prevent an acute rise of
• Diuretics (unless patient is fl
fluid overloaded) serum creatinine after intravenous contrast, but
• Low-dose dopamine survival and need for dialysis were not affected.

Vasopressor Agents and the Kidney Tight Glucose Control


Vasopressor agents are often needed to manage A single-center randomized controlled trial on
septic shock, and noradrenaline and dopamine are intensive insulin therapy in postoperative
good first-line drugs. Although there are few direct ventilated patients showed a 41% decrease in the
comparison studies, patients with septic shock incidence of ARF requiring dialysis in the group
tend to respond better to noradrenaline. Concern of patients whose blood sugars were tightly con-
regarding the potential for noradrenaline to impair trolled between 4.4 and 6.1 mmol/L compared
renal and mesenteric perfusion has been reduced with patients whose blood sugar was allowed to
by studies showing that reversal of hypotension rise to 12 mmol/L before insulin was initiated.
with noradrenaline outweighed this effect and Further studies are necessary to confirm
fi these
increased renal and mesenteric perfusion. results.

Prevention Prophylactic Dopamine or Diuretics


A recent meta-analysis of 61 trials showed that
General Vigilance low-dose dopamine (<5 μg/kg/min) often increased
Meticulous attention to fluid balance, blood pres- urine output but had no effect on renal function
sure, prescribed drugs, and treatment of sepsis are or prevention of ARF.
the most important preventive measures. There is
insuffi
ficient evidence to recommend specifi fic phys-
Natriuretic Peptides
iological targets (MAP, cardiac output, filling pres-
sures) that will ensure adequate renal perfusion. Urodilatin (renal natriuretic peptide) or anaritide
Instead, therapy needs to be individualized based (synthetic form of atrial natriuretic peptide)
on the baseline physiological condition of the have failed to show any protective effect on the
individual patient. kidney.
6. Acute Renal Failure in the Surgical Patient 37

Future Advances ing open heart surgery: risk factors and prognosis.
Perfusion 2005; 20: 317–322.
2. Barrett BJ, Parfrey PS. Preventing nephropathy
At present, no agents have conclusively demon-
induced by contrast medium. N Engl J Med d 2006; 354:
strated a protective effect against ARF or altera- 379–386.
tion of the course of ARF. Strategies aimed at 3. Chertow GM, Levy EM, Hammermeister KE, Grover
modulating renal function and renal recovery F, Daley J. Independent association between acute
have focused on several mechanisms: renal failure and mortality following cardiac surgery.
Am J Med d 1998; 104: 343–348.
1. Reduction of renal metabolism and energy
4. Friedrich JO, Adhikari N, Herridge MS, Beyene J.
consumption of the kidneys (i.e., induction of Meta-analysis: low-dose dopamine increases urine
hypothermia, use of insulin-like factor I). output but does not prevent renal dysfunction or
2. Modulation of the inflammatory
fl system (i.e., death. Ann Intern Med d 2005; 142: 510–524.
up-regulation of the acute stress response, 5. Lassnigg A, Donner E, Grubhofer G, Presterl E, Druml
manipulation of complement system, blockade W, Hiesmayr M. Lack of renoprotective effects of
of adhesion molecules). dopamine and furosemide during cardiac surgery.
3. Ischemic preconditioning. J Am Soc Nephroll 2000; 11: 97–104.
6. Mangano DT, Tudor IC, Dietzel C. Multicenter Study
These strategies are clearly important areas of of Perioperative Ischemia Research Group; Ischemia
research but not ready for clinical application. Research and Education Foundation. The risk associ-
ated with aprotinin in cardiac surgery. N Engl J Med
References 2006; 354: 353–365.
7. Van den Berghe G, Wouters PJ, Bouillon R, et al.
1. Bahar I, Akgul A, Ozatik MA, Vural KM, Demirbag Intensive insulin therapy in critically ill patients.
AE, Boran M, Tasdemir O. Acute renal failure follow- N Engl J Med d 2001; 345: 1359–1367.
7
Rhabdomyolysis and
Compartment Syndrome
Laurie Tomlinson and Stephen Holt

Rhabdomyolysis occurs when an insult causing necrosis. There may be severe pain, with loss of
myocyte necrosis results in release of intra- muscle function and loss of distal pulses. The
cellular contents into the circulation. Renal dys- diagnosis may be occult, especially in the uncon-
function is caused by a combination of renal scious patient. Direct pressure measurements
vasoconstriction, tubular damage, and tubular can be made by passing a needle connected to a
obstruction. pressure manometer (e.g., central venous pressure
[CVP] transducer) into the affected muscle
compartment.
Causes A fasciotomy should be considered if the pressure
exceeds 40 mmHg or greater than 30 mmHg above
Rhabdomyolysis accounts for approximately 7% diastolic pressure.
of all causes of acute renal failure (ARF) during
peacetime. This figure is much higher after natural
disasters and in wartime. For example, after the Diagnosis
1998 Turkish earthquake, 12% of the hospitalized
population developed significant
fi renal dysfunc- Serum changes consequent on rhabdomyolysis:
tion and 477 patients required dialysis.
Direct crush or compression injury and drugs
are the most important causes in clinical practice, Creatine Kinase
see Table 7.1. There are often predisposing factors,
for example, alcohol, which can presensitize myo- Very high levels of the muscle enzyme CK are
cytes so they may be damaged by a more trivial pathognomic of this condition. The degree of eleva-
insult. A clinical scoring system exists (not widely tion is proportional to the degree of muscle injury.
used) based on levels of phosphate, potassium, Other muscle enzymes, such as aspartate transami-
albumin, creatine kinase (CK), and presence of nase (AST) and lactate dehydrogenase (LDH) are
dehydration and sepsis. also elevated. CK levels should decline by approxi-
mately 40% per day, a plateau or an increase should
prompt a search for ongoing muscle damage.
Compartment Syndrome
Hyperkalemia
After an appropriate precipitant, inflammation

within a muscular compartment causes a vicious Hyperkalemia caused by efflux
fl of potassium from
cycle of increasing pressure. This leads to further damaged cells is an early and life-threatening
infl
flammation and damage, eventually compro- consequence of rhabdomyolysis. It should be
mising blood supply, leading to further muscle aggressively treated.

38
7. Rhabdomyolysis and Compartment Syndrome 39

TABLE 7.1. Causes of rhabdomyolysis


Physical Trauma, hyperthermia, hypothermia, exercise, electric shock, seizures, delirium tremens
Toxins and drugs Alcohol, statins, amphetamines, aspirin (overdose), barium, barbiturates, caffeine, carbon monoxide, ecstasy, ethylene
glycol, LSD, malignant hyperpyrexia, neuroleptic malignant syndrome, opiates, toluene, snake/insect bites, vasopressin
Muscle ischemia Vascular ischemia, coma, sickle cell disease, surgery, vasoconstrictors, CO2 angiography
Infection Virtually any viral or bacterial infection (e.g., influenza, HIV, Epstein-Barr virus, Legionella, tetanus, malaria, Bacillus cereus)
Metabolic Hypernatremia/hyponatremia, hypokalemia, hypophosphatemia, diabetic ketoacidosis, diabetic hyperosmolar coma,
water intoxication, myxedema
Inherited Deficiency of carnitine palmityl transferase II, phosphofructokinase, myophosphorylase (McArdles), myoadenylate deaminase,
cytochrome oxidase, succinic dehydrogenase, coenzyme Q10 deficiency, King-Denborough Syndrome, Wilson’s disease
Immune Polymyositis, dermatomyositis

Acidosis rise in parathyroid hormone (a reflex


fl to the initial
hypocalcemia).
Metabolic acidosis may be caused by increased Symptoms of hypocalcemia are rare and treat-
lactate production and lactate release by damaged ment with intravenous calcium should be avoided
muscle. Myoglobin (Mb) is considerably more unless tetany or cardiac dysfunction is present.
toxic in an acid milieu. Pharmacologically administered calcium is taken
up avidly by the damaged muscle. It may be depos-
Early Hypocalcemia and Late Hypercalcemia ited as inorganic complexes causing “heterotopic
calcification,”
fi which delays recovery and can lead
Serum calcium levels often fall dramatically, to long-term muscle dysfunction.
with total calcium less than 1.7 mmol/L in the
early stages. This is caused by sequestration into
damaged muscle and reperfusion-induced cellu- Hyperphosphatemia
lar calcium uptake. In contrast, intracellular Serum phosphate often exceeds 3 mmol/L.
calcium concentrations in damaged muscle may
rise by up to 10-fold. In the recovery phase of
rhabdomyolysis, serum calcium levels normalize Urinary Abnormalities
and may even “overshoot,” secondary to calcium Urine dipsticks are usually positive for blood
release by recovering myocytes and a transient because they detect the heme moiety present in
both hemoglobin (Hb) and Mb. On microscopy,
few red cells are seen (unless there is coexistent
trauma), instead, the characteristic “brown sugar
casts” of Mb are seen (Figure 7.1). When there is
uncertainty, Mb can be specifically
fi assayed in the
urine, although it has a short half-life. An assay
exists for myosin heavy chain, which remains pos-
itive for up to 12 days after the initial insult.

Pathophysiology of ARF
Suggested causes:
1. A reduction in renal blood flow
fl . There is a
FIGURE 7.1. “Brown sugar” Mb casts under light microscope are reduction in the effective blood volume caused
similar to granular casts but have a brown/rusty tinge. Additional by fluid
fl shifts from the intravascular to extracel-
red cells, tubular cells, and other debris are also present within lular fluid
fl compartments. Mb binds to nitric
the urine. oxide (NO), preventing intrarenal vasodilation
40 L. Tomlinson and S. Holt

(especially in the medulla) and, in addition, vaso- • Bicarbonate solutions that are more concentrated
dilators (e.g., endothelin) are increased. can be administered in small aliquots, e.g., 50 mL
2. Direct heme protein tubulotoxicityy occurs, of 8.4% NaHC03 via central access in patients
probably by free radical-mediated mechanisms. who are intravascularly full—remembering that
3. Tubular cast formation. Urinary Mb and this is 1 mmol of sodium per milliliter of fluid

Tamm-Horsfall protein (THP) complex and precip- and may cause sodium/fluid
fl overload
itate as tubular casts. These casts are less soluble in
acidic conditions. Although there is some evidence
Mannitol
that these complexes cause tubular obstruction,
micropuncture studies have shown relatively low Mannitol promotes an osmotic diuresis and may
intratubular pressures, suggesting that these casts reduce pressure in a swollen muscle compartment,
are as a result of reduced tubular flflow and reduced but it also causes osmotically induced tubular
washout rather than by obstruction per se. damage with vacuolation. There is no good evi-
dence that it is more effective than saline alone
and it has little scientifi
fic rationale to recommend
Treatment its routine use.

Intravascular Volume Expansion Dialysis/Hemofiltration


Intravascular volume expansion at the first
fi possi-
The circulating concentration of Mb can be
ble opportunity after the insult is the single most
reduced by hemofi filtration, plasma exchange, and
effective therapeutic maneuver in rhabdomyoly-
hemodialysis, with dialysis being somewhat less
sis. This not only prevents or limits renal damage
successful. It has not been shown that any physical
but may play a role in preventing acidosis and
therapy materially reduces renal Mb burden or
limiting ongoing damage caused by hypoperfu-
shortens the duration of renal replacement
sion. Very large volumes of fluid
fl can be lost into
therapy. Physical therapies may have a role, if
areas of muscle injury. In trauma situations in
commenced early, or if Mb release can be antici-
which there is a risk of crush injury, fl
fluid resusci-
pated, such as during arterial surgery.
tation should be commenced before the victim is
extricated.
Prognosis
Alkalinization
There is an unquantifiable
fi early mortality, mainly
There is much compelling evidence to suggest that
caused by hyperkalemia or the insult. Mortality
urinary alkalinization greatly reduces the nephro-
after diagnosis is up to 20%, usually caused by
toxicity of Mb. However, there are no large human
other associated conditions, e.g., sepsis. Survivors
trials that confirm
fi this consistent finding from
of the Japanese earthquake in 1995 arriving in
animal research. The potential benefits fi in this
hospital within 6 hours had an approximately 20%
setting include reduced renal vasoconstriction, a
chance of developing ARF, whereas all of those
dramatic reduction in the ability of Mb to cause
arriving after 40 hours developed renal failure. If
oxidant damage, and increased solubility of Mb-
the patient recovers from the initial insult, the
THP complex.
renal dysfunction almost always resolves, but can
Alkalinization can lower ionized calcium still
take up to 3 months.
further and, if administered, it is wise to periodi-
cally check the ionized calcium.
A suggested fluid
fl replacement regime would
be:
Summary
• Isotonic bicarbonate (1.26% sodium bicarbonate) • Rhabdomyolysis is a common cause of ARF, with
until urine pH is greater than 7 if the patient is important early biochemical changes that may
intravascularly volume deplete (which is usual) be fatal if treatment is not instituted quickly
7. Rhabdomyolysis and Compartment Syndrome 41

• Compartment syndromes can be occult and are Suggested Reading


important to detect and monitor to protect
against further renal injury Holt SG, Moore KP. Pathogenesis and treatment of renal
• Early treatment with volume replacement dysfunction in rhabdomyolysis. Intensive Care Med.
(±alkalinization) will reduce the risk of renal 2001 May; 27(5): 803–811.
failure and the need for renal replacement Zager R. Rhabdomyolysis and myohemoglobinuric acute
therapy renal failure. Kidney Int. 1996 Feb; 49(2): 314–326.
8
Multisystem Causes of Acute
Renal Failure
Tim Leach

This chapter covers some of the more specialized Normally, the immune system surveys cells and
causes of acute renal failure, which, although more tissues within the body, recognizing and ignoring
likely to present to the nephrologist, could be cells expressing “self” antigens while attacking
admitted to the intensive care unit as a conse- cells without these protective epitopes. In autoim-
quence of their illness or because of complications mune conditions, the immune system does not
of their treatment. protect cells with “self” expression. Cells are
attacked and either inflflamed or killed, or circulat-
ing self-antigens are bound with antibody-forming
Systemic Vasculitis immune complexes. Immune complexes are very
large molecules that are often unable to pass
Vasculitis is the term given to inflammation
fl of through capillaries because of their size. They can
blood vessels. Vasculitis is a rare condition, with induce local inflflammation and activate the com-
an incidence of approximately 6 people per million plement cascade.
(Western) population per year (1). Vessels can be
classified
fi according to their size (Table 8.1) (2).
Renal failure can occur in any vasculitis, but this
chapter focuses on those conditions that affect
Presentation
renal function directly through inflammation

Symptoms
within the glomeruli (small vessels), rather than
affecting the kidneys indirectly through a reduc- Renal failure as a result of fulminant small vessel
tion of blood supply to the kidneys (large and vasculitis will present acutely, and patients may
medium vessel diseases). be systemically unwell, requiring organ support.
More often, however, there is an indolent presenta-
tion with several months of nonspecificfi symptoms
Etiology and signs (Table 8.2).

Small vessel vasculitides separate into two broad


groups: those in which immune complexes are Signs
deposited within the renal glomeruli and those
Fulminant systemic vasculitis presents with the
without evidence of deposition histologically. The
nephritic syndrome:
latter group are termed pauci-immune; lacking
(literally “few”) immune complexes. This distinc- • Azotemia (more often with oligoanuria)
tion is useful for making a histological diagnosis • Hypertension and edema from fl fluid overload
and for estimating prognosis, but the underlying (but can present with circulatory collapse caused
cause of these small vessel vasculitides is similar: by vasodilation and dehydration)
autoimmunity. • Hematuria with red cell casts

42
8. Multisystem Causes of Acute Renal Failure 43

TABLE 8.1. Vasculitic conditions according to the vessel size


affected
Vessel size Condition
Large Takayasu’s arteritis
Giant cell arteritis
Medium Polyarteritis nodosa
Kawasaki disease
Small Pauci-immune:
• Wegener’s granulomatosis (WG)
• Microscopic polyangiitis (MPA)
• Churg-Strauss disease (CSD) Immune-complex
forming
• Goodpasture syndrome (GS) Immune-complex
depositing:
• SLE
• Henoch-Schönlein purpura (HSP)
• Cryoglobulins
• Rheumatoid arthritis FIGURE 8.1. Plain chest x-ray of fulminant pulmonary hemorrhage
of 17-year-old male patient with systemic vasculitis caused by
Source: Jeanette et al., 1994. Henoch-Schönlein purpura. The patchy diffuse alveolar shadowing
is clearly seen.
Other features include:
pneumonitis (Figure 8.1), or superadded
• Fever pneumonia
• Raised purpuric rash
• Respiratory failure secondary to pulmonary
edema, pulmonary hemorrhage from vasculitic Laboratory Investigations (Table 8.3)
TABLE 8.2. Nonrenal symptoms suffered in systemic vasculitis Imaging
Relative frequency Chest x-ray may be clear or show pulmonary
(some patients
edema or pulmonary hemorrhage (Figure 8.1).
System experienced more
affected than one) Symptoms Renal ultrasound will usually show normal-sized
kidneys with no obstruction.
Lower 63% Cough
respiratory Breathlessness (on
exertion/at Renal Biopsy
rest/orthopnea)
Hemoptysis The ultimate investigation for renal vasculitis is
Upper 50% Sinusitis the biopsy. Patients need to be hemodynamically
respiratory Nasal congestion
stable, normotensive, have a normal platelet count
Reduced hearing
Epistaxis and coagulation screen and be able to lie flat
fl (see
Skeletal 42% Arthralgia Figure 8.2).
Arthritis
Synovitis
Muscular 33% Myalgia
Myositis
Treatment
Muscle weakness
Dermatological 22% Rash (purpura/echinoses/ Treatments of patients with vasculitis fall into
malar flush) three main areas:
Neurological 14% Headache
Lethargy
Loss of concentration Resuscitation
Coma
Patients should be stabilized in terms of airway,
Source: Hedger et al., 2000. breathing, and circulation, as in any serious illness.
44 T. Leach

TABLE 8.3. Investigations for systemic vasculitis


Investigation Rationale Expected result
Urea and electrolytes Likely renal failure Elevated urea and creatinine
Potentially elevated potassium
Full blood count Anemia Anemia
Elevated white cell count
Normal/elevated platelet count (cff HUS)
Antineutrophil cytoplasmic antibody WG and MPA associated with positive ANCA Positive cANCA in WG
(ANCA) Positive pANCA in MPA
Otherwise negative
Antiglomerular basement antibody Goodpasture syndrome Positive in GS
(aGBM) Otherwise negative
Complement Involved in inflammation Usually mildly or significantly reduced levels of
C3 and C4
ESR/CRP Inflammatory markers Usually significantly elevated

ESR, erythrocyte sedimentation rate; CRP, C-reactive protein.

Maintenance Disease-Specific Treatment


Maintenance of adequate oxygenation, circula- The disease is one of an abnormal immune
tion, and fluid balance is vital to their recovery. response inducing renal and other organ inflam-

Hemofi filtration, hemodiafi filtration, or hemodialy- mation and treatment falls into three groups.
sis depending on the patient’s hemodynamic
1. Reduction of inflammation
fl with corticoste-
stability should be used to prevent and treat com-
roids (e.g., intravenous methylprednisolone fol-
plications that do not respond to standard medical
lowed by oral prednisolone).
therapies. Infection, bleeding, and malnutrition
2. Reduction of antibody formation with
should be sought and remedied.
immunosuppressive agents (e.g., cyclophospha-
mide administered intravenously every 2 to 4
weeks or orally everyday).
3. Removal of the already-formed antibodies
with therapeutic plasma exchange if the patient has
pulmonary hemorrhage (potentially life threaten-
ing) or is requiring renal dialysis (severe disease).

Outcome
Renal vasculitis is a serious illness with significant

morbidity and mortality. Before treatment, it was
universally fatal; with treatment, mortality is 10%
at 18 months. Up to 50% of patients require dialy-
sis with no renal recovery. If the patient did not
require dialysis at presentation, there is a 91%
FIGURE 8.2. Photomicrograph of renal biopsy from patient with
Wegener’s (cANCA-positive) renal vasculitis. Hematoxylin and
chance of renal survival. If dialysis dependent at
eosin stain with silver counterstain, showing features consistent presentation, 30% of patients may regain renal
with the disease. The glomerulus (black arrow)
w shows segmental function with cyclophosphamide and corticoste-
fibrinoid change (red arrows) and crescent formation (yellow
( roids alone, but 90% of patients may develop renal
arrow).
w Some tubules contain red cells (white arrows). (Photo recovery with the addition of therapeutic plasma
reproduced with the kind permission of Dr. Nicholas Marley.) exchange (3, 4).
8. Multisystem Causes of Acute Renal Failure 45

Infection is the most significant


fi side effect of
treatment. Bacterial sepsis, viral infections such as
herpes zoster and cytomegalovirus, fungal sepsis,
and Pneumocystis carinii pneumonitis (PCP)
occur in 40% of treated patients (5). Prophylaxis
against fungi and PCP is recommended.

Systemic Lupus Erythematosus


Systemic lupus erythematosus (SLE) is a multisys-
tem autoimmune condition of primarily young
(20–30 years old) women (Figure 8.3). The Ameri-
can College of Rheumatology definesfi the diagno-
sis of lupus for a patient presenting with four or
more of the following features at the same time, or
individually during a period of time:
1. Malar rash: redness or rash that may appear
in a butterflfly confi figuration across the nose
and cheeks. It can appear on one or both sides
of the face and is usually flat.
fl FIGURE 8.3. Magnetic resonance imaging scan of brain of 21-year-
2. Discoid rash: thick raised patches that can old woman with cerebral lupus. T2-weighted sagittal section
occur on any part of the body and may result shows increased white and grey matter signal, particularly in the
frontal and parietal regions, in keeping with cerebral vasculitis.
in scarring.
3. Sun sensitivity: a reaction to sunlight that is
more severe than just sunburn. interstitial pneumonitis, or aseptic meningitis. A
4. Oral ulcers: frequent development of mouth positive Coombs test, low complement levels, and
or nose ulcers. immune deposits at the dermal-epidermal junc-
5. Arthritis: pain, tenderness, or swelling in two tion on skin biopsy are also suggestive of lupus.
or more joints. Patients with SLE can present in extremis with
6. Pleurisy or pericarditis. any system involvement. The presence of a multi-
7. Nephritis: proteinuria or cellular casts in the system disease such as SLE should always be
urine. entertained in such patients.
8. Nervous system disorder: seizures or psy-
chotic behavior that cannot be attributed to
Laboratory Testing
drugs or metabolic dysfunction.
9. Blood system disorder: hemolytic anemia, leu- Tests that provide potentially diagnostically useful
kopenia, lymphopenia, or thrombocytopenia. information when SLE is suspected include:
10. Immunologic disorder: the presence of the
• Complete blood count and differential
lupus erythematosus (LE) cell, a false-positive
• Serum creatinine
reaction to the tests for syphilis, or the pre-
• Serum albumin
sence of autoantibodies.
• Serologic test for syphilis (falsely positive
11. Positive antinuclear antibodies (ANA): anti-
because of cross-reactivity)
bodies against the nucleus of cells, particu-
• Urinalysis
larly against double-stranded DNA.
• 24-hour urine collection for calculation of creati-
Lupus should also be suspected in young nine clearance and quantifification of proteinuria
women presenting with purpura, easy bruising, • Autoantibody testing: ANA, antibodies to phos-
diffuse lymphadenopathy, hepatosplenomegaly, pholipids, antibodies to double-stranded DNA,
peripheral neuropathy, endocarditis, myocarditis, and antibodies to Smith (Sm)
46 T. Leach

• Complement levels (total hemolytic comple- Thrombotic Microangiopathy


ment [CH50], C3 and C4)
Thrombotic microangiopathy (TMA) covers the
acute syndrome of microangiopathic hemolytic
Imaging
anemia, thrombocytopenia, and variable signs of
This may be valuable but is not routinely obtained organ injury caused by platelet thromboses in
unless indicated by the presence of symptoms, the microcirculation. Two clinically distinct but
clinical findings, or laboratory abnormalities. pathologically identical syndromes are described:
Examples include: hemolytic uremic syndrome (HUS) and throm-
botic thrombocytopenic purpura (TTP). HUS
• Plain radiographs of involved joints
usually affects children with renal failure but
• Renal ultrasound to assess kidney size and rule
minimal neurological involvement. TTP is a
out an obstructive post-renal cause when there
disease of adults, with predominantly neurologi-
is evidence of acute renal failure
cal involvement and variable other organ disease.
• Chest X ray
The two syndromes often overlap and, thus, are
• Echocardiography (for suspected pericardial
termed HUS/TTP.
involvement or to seek a source of emboli)
• CT scanning (for abdominal pain, suspected
pancreatitis, lymphadenopathy) Etiology
• Magnetic resonance imaging (for seizure activ- The mechanisms behind the development of
ity, focal neurologic defificits or cognitive dys- TMA are poorly understood but seem to involve
function/personality changes—see Figure 8.3) endothelial injury, which triggers events leading
• Contrast angiography may be valuable if vascu- to microvascular thrombosis, microangiopathic
litis affecting medium-sized arteries is suspected hemolytic anemia, and platelet consumption.
(mesenteric or limb-threatening ischemia). Most causative factors lead to toxicity of the
endothelial cells: autoantibodies, exotoxins and
Treatment endotoxins, immune complexes, and certain
drugs.
Treatment is broad and depends on the system The pathology of TMA consists of capillary and
involved. General principles are: arteriolar wall widening, with swelling and detach-
• Reduce inflammation
fl with corticosteroids ment of the endothelium, and occlusion or severe
• Prevent further autoantibody production with restriction of the lumen and platelet micro-
immunosuppressant medications such as cyclo- thrombi. In HUS, this occurs mainly in the kidney,
phosphamide or mycophenolate mofetil in TTP, mainly in the brain.
• Remove circulating autoantibodies with thera-
peutic plasma exchange Presentation
Antiphospholipid antibodies and the presence HUS/TTP rarely causes specifi fic symptoms. In the
of the lupus anticoagulant increase coagulation situations shown in Table 8.4, vomiting, pallor,
and lead to arterial and venous thromboses. Use purpura, anuria, and/or neurological signs should
of antiplatelet drugs, thrombolytics, and antico- alert the clinician to the possibility of TMA.
agulation in these patients often prevents further
problems.
Differential Diagnosis
Fertility and pregnancy in lupus patients are
often problematic. Pregnancy alters the immune • Systemic vasculitis will usually present with
state, often leading to fl
flares of lupus postpartum. arthralgia/arthritis, and the platelet count will
Coagulation abnormalities often lead to difficul-
fi be normal and will rarely have central neuro-
ties with conception and spontaneous abortion. logical involvement.
Primary infertility or recurrent miscarriages are • Disseminated intravascular coagulation (DIC)
relatively common presenting features of SLE. is usually associated with shock or obstetric
8. Multisystem Causes of Acute Renal Failure 47

TABLE 8.4. Conditions and situations associated with the development of


HUS/TTP
Acquired Shigatoxin (Escherichia colii 0157)
Pregnancy
Pneumococcal infection
Systemic disease HIV infection
SLE
Scleroderma
Malignancy
Drug associated Mitomycin C, tamoxifen,
bleomycin, cisplatin,
clopidogrel, quinine, interferon,
OKT3, cyclosporine, tacrolimus
(among others)
Organ transplant De novo (usually drugs)
Recurrent posttransplant HUS
Genetic and familial forms
Idiopathic and atypical forms

complications and will have consumption of all patients can die in the acute phase. Cerebrovascular
of the clotting factors. accidents, seizures, and coma occur in 25% of
• Malignant hypertension will have classical patients, and residual impairment of renal
retinal changes, signifificant high blood pressure excretory function is present in up to 40% of
(usually >210/130), and usually a history of patients.
hypertension. HUS/TTP in adults or children older than 14
years old usually requires treatment. In those with
an apparent cause (pregnancy, malignancy, infec-
Laboratory Investigations tion, or drugs), removal of the cause is essential.
• Low hemoglobin (<7 g/dL) Supportive treatment is required. Specific fi treat-
• Thrombocytopenia (<80 × 109 cells/L) ment of the condition revolves around therapeutic
• The blood filmfi will show red cell fragments plasma exchange or plasma infusion if TPE is
(schistocytes) and increased reticulocyte not available. There is no evidence of benefi fit of
counts immunosuppression with corticosteroids, immu-
• Elevated lactate dehydrogenase and indirect noglobulins, or vincristine, or of benefit fi from
bilirubin (caused by hemolysis) antithrombotic or antiplatelet agents.
• Haptoglobin levels will usually be low because Rescue therapies from severe refractory or
of consumption relapsing disease include bilateral nephrectomy
• Coombs test is negative and/or splenectomy.
• Moderate proteinuria (1–2 g/d) with few red
cells and casts (ARF is secondary to occlusion of
capillaries rather than inflammation)
fl References
1. Hedger N, Stevens J, Drey N, Walker S, Roderick P.
Treatment and Outcome Incidence and outcome of pauci-immune rapidly
progressive glomerulonephritis in Wessex, UK: a 10-
The epidemic or sporadic diarrhea-associated year retrospective study. Nephrol Dial Transplant
HUS/TTP of young children is usually self-limiting 2000; 15(10): 1539–1539.
and mild. Renal failure requires dialysis in approxi- 2. Jennette JC, Falk RJ, Andrassy K. Nomenclature of
mately 50% of patients, but otherwise supportive systemic vasculitides. Proposal of an international
treatment is all that is required. Ninety percent of consensus conference. Arthritis Rheum 1994; 37:
patients should recover completely. Up to 5% of 187–192.
48 T. Leach

3. Andrassy K, Kuster S, Waldherr R, Ritz E. Rapidly pro- lonephritis without anti-GBM antibodies. Kidney Int
gressive glomerulonephritis: analysis of prevalence 1991; 40(4): 757–763.
and clinical course. Nephron 1991; 59(2): 206–212. 5. Hoffman GS, Kerr GS, Leavitt RY, et al. Wegener gran-
4. Pusey CD, Rees AJ, Evans DJ, Peter DK, Lockwood ulomatosis: an analysis of 158 patients. Ann Intern
CM. Plasma exchange in focal necrotizing glomeru- Medd 1992; 116: 488–498.
9
Therapeutic Plasma Exchange
Tim Leach

Therapeutic plasma exchange (TPE) is an extra- • Of molecular weight greater than 15,000 kDa
corporeal blood purifi fication technique designed so it cannot be removed in any other way,
for the removal of large molecular weight sub- and/or
stances from plasma. Large molecular weight • Of suffi
ficient half-life that TPE is quicker than
substances equilibrate slowly between the vascu- endogenous removal, and/or
lar space and the interstitium. Calculations of • Acutely toxic and resistant to conventional
the rate of their removal by TPE follows fi first- therapy
order kinetics, i.e., approximately 60% is removed
by a single plasma volume exchange, and 75%
by an exchange equal to 1.4 times the plasma
volume.
Prescription (Table 9.1)
Blood is pumped through a highly permeable
Calculation of plasma volume:
filter, replacing the filtrate with fluid as indicated

(Table 9.1). Venous access on the intensive care Estimated plasma volume (L) = 0.07 × Weight
unit (ICU) is via a double-lumen dialysis catheter, (kg) × (1 − hematocrit)
but can be via two wide-gauge peripheral venous
1. Before each treatment, measure serum potas-
cannulae. If chronic therapy is indicated, an arte-
sium calcium and clotting screen.
riovenous fifistula is used. The patient and filter are
2. Calculate the estimated plasma volume: Volume
anticoagulated during the procedure.
of exchange is measured in Patient Plasma
Volumes (∼3 L).
3. Prescribe the plasma exchange:
Indications (Table 9.2) a. Number and spacing of treatments
b. Volume and type of fl fluid
The basic premise of TPE is that removal of large
4. Electrolyte supplementation as needed (potas-
molecular weight substances from the circulation
sium and calcium).
will reduce further damage and may permit rever-
5. If coagulopathic consider, fresh-frozen plasma
sal of the pathological process.
(FFP) as the final exchange volume.
Other benefits
fi include unloading the reticulo-
endothelial system to permit further endogenous
removal of circulating toxins, stimulation of lym-
After Procedure
phocyte clones, and allowing re-infusion of large
volumes of plasma without the risk of volume • Repeat electrolytes and clotting after 2 hours
overload. and increase supplementation as needed
For TPE to be appropriate, the substance to be • FFP can be given as the final exchange volume
removed should be: if coagulopathic

49
50 T. Leach

TABLE 9.1. Example of TPE prescriptiona


Indication Plasma volumes Replacement fluid Number of exchanges Exchange frequency
Rapidly progressive 1 to 1.5 Albumin, with FFP if 7 to 10 Daily or alternate days
glomerulonephritis pulmonary hemorrhage
Acute renal failure caused
by myeloma kidney
Hyperviscosity 1 Albumin/saline mixtureb Until symptoms subside or Daily
Syndrome plasma viscosity
normal
Anti-glomerular basement 1.5 Albumin, with FFP if 7 to 10 Daily
membrane (GBM) disease pulmonary hemorrhage
Hemolytic uremic 1 to 2 All FFP Until platelets normal/no Once or twice daily
syndrome/thrombotic red cell fragments
thrombocytopenic purpura (usually 7 to 16)
Guillain-Barré Syndrome 2 Albumin 4 Alternate days
a
From: Wessex Renal and Transplant Unit, United Kingdom.
No more than one part saline to two parts albumin (i.e., ≤1 L saline for 2 L albumin).
b

TABLE 9.2. American Association of Blood Banks indications for Complications


TPE
1. Standard and acceptable 2. Sufficient evidence to suggest • Hypotension: vasovagal, hypovolemia
• Chronic inflammatory efficacy/acceptable adjunct • Fluid overload
demyelinating • Cold agglutinin disease
• Hypocalcemia causing tetany
polyneuropathy • Protein-bound toxins
• Cryoglobulinemia (drug overdose/ • Hypokalemia
• Anti-GBM disease poisonings) • Coagulopathy caused by removal of clotting
• Guillain-Barré syndrome • Hemolytic uremic factors or thrombocytopenia with heparin
• Familial hypercholesterolemia syndrome anticoagulation
• Myasthenia gravis • Rapidly progressive
• Protein-bound drug removal (administer drugs
• Posttransfusion purpura glomerulonephritis
• Thrombotic • Systemic vasculitis afterr TPE)
thrombocytopenic purpura • Acute renal failure caused • ACE inhibitors may cause fl flushing and hypoten-
by myeloma kidney sion (stop 24 h before treatment)
3. Inconclusive evidence orr 4. No efficacy in trials
uncertain benefit-to-risk ratio • AIDS
• ABO-incompatible organ or • Amyotrophic lateral
marrow transplantation sclerosis
• Coagulation factor inhibitors • Dermato/polymyositis
• Idiopathic thrombocytopenic • Psoriasis
purpura • Renal transplant rejection
• Multiple sclerosis • Rheumatoid arthritis
• Progressive systemic sclerosis • Schizophrenia
• Thyroid storm
• Warm autoimmune
hemolytic anemia
10
Renal Replacement Therapy
John H. Reeves

Acute renal failure (ARF) occurs in 7% of patients maintain a concentration gradient favoring the
admitted to the intensive care unit (ICU) (1). Pre- out-diffusion of unwanted solutes. Both dialysate
viously, mortality exceeded 91%, but with the and replacement solutions contain electrolytes in
introduction of dialysis, this quickly fell to approx- physiological concentrations and some form of
imately 50% (2). Overall mortality for ARF has buffer base.
remained approximately 50%, associated with The hemofilter
fi r or dialysis membrane’s permea-
increasing comorbidity (3). bility to water is determined by its surface area
There is no specifi
fic therapy for ARF other than (typically 0.5 to 2.0 m2) and the number and size
removal of the cause and ongoing supportive care of its pores (typically 0.0055-μm diameter in a
awaiting spontaneous recovery. Renal replacement high-flux
fl hemofi filter). The pore size determines
therapy (RRT) is the cornerstone of that support- the size of molecules that are freely fi
filtered along
ive care. with water. Current hemofilters
fi freely filter sub-
stances up to approximately 5000-D molecular
weight and then in decreasing amounts up to a cut
The Basics off of approximately 20,000 Da. This minimizes
loss of important larger molecules, such as
Extracorporeal RRT involves the passage of a albumin (molecular weight, 57,000 Da).
patient’s blood outside his/her body through a The ratio between the concentration of solute
dialysis or hemofilter
fi machine, where the removal in filtrate and that in plasma water is called the
of unwanted solutes and excess water and the sieving coefficient
fi , and this concept becomes
replacement of lost bicarbonate (or buffer base) important in calculating the clearance of interme-
take place. The “purified”
fi blood is returned to the diate-sized molecules. The sieving coefficient
fi for
patient. a small unbound solutes is one, decreasing to zero
Clearance can be definedfi as that volume of as molecular size and or plasma protein binding
plasma completely cleared of a substance in a increases.
given time. During extracorporeal RRT, net solute Theoretical small solute clearance can be pre-
clearance can be achieved by ultrafiltration
fi across dicted by knowledge of the blood flow
fl through the
a porous membrane down a pressure gradient (fi (fil- extracorporeal circuit (QB) and the rate of ultrafil-

tration), or by diffusion across a semipermeable tration (QF) and dialysate flow
fl (QD) (Figure 10.1).
membrane down a concentration gradient (dialy- During continuous RRT (CRRT), when blood
sis), or both. During fifiltration, the filtrate is dis- flow is signifi
fl ficantly higher than dialysate or ultra-
carded and a replacement solution is added to the filtration rates, small solute clearance is deter-

blood to maintain fluid
fl and electrolyte equilib- mined by dialysate and ultrafiltrate
fi flow. Assuming
rium. During dialysis, a continuous stream of complete concentration equilibrium between dial-
dialysate is passed in the opposite direction to ysate and plasma water and a sieving coefficient

blood, on the nonblood side of the membrane, to equal to one:

51
52 J.H. Reeves

Haemofiltration replacement ≅ QF week. In contrast, ICU patients with ARF may


require daily treatment (7).
Blood flow Hybrid: there is increasing interest in hybrid
Membrane QB approaches for extracorporeal RRT in the ICU
(8, 9). For example, during sustained low-
efficiency
fi dialysis (SLED), daily intermittent
therapy is reduced in intensity and extended in
duration up to 8 or 12 hours. The regular breaks
Effluent Dialysate aid staffi
fing and free patients for investigations
QE QD and procedures. The reduced intensity reduces
the cardiorespiratory destabilization associated
FIGURE 10.1. Predicting clearance during RRT.
with IHD.

Clearance during CRRT = QF + QD = QE Access


In contrast, during intermittentt hemodialysis Arteriovenous: arteriovenous access involves can-
(IHD), dialysate flow is signifificantly higher than nulation of a medium-sized artery and large
blood flow, and blood flow becomes the limiting vein—often the femoral artery and vein. Blood
factor for solute clearance. Small solute clearance flows passively from the artery through the
is proportional to the plasma flowfl through the extracorporeal circuit (ECC) and back through
dialyzer: the vein, driven by the mean arterial pressure.
This method is reserved for situations in which
Clearance during IHD ≅ QB × (1 − hematocrit) resources are limited.
This simplifified analysis only holds for small, Venovenous: venovenous access involves cannula-
unbound solutes, such as urea and creatinine. tion of central veins, most commonly with a
Increasing molecular weight decreases diffusive double-lumen cannula in the femoral, internal
clearance more signifi
ficantly than convective clear- jugular, or subclavian vein. Blood flow
fl is driven
ance. Binding to macromolecules, such as albumin, by a pump in the ECC. This increases reliability
decreases convective and diffusive clearance (4). of blood flow
fl and maximizes solute clearance,
but introduces the need for more complex safety
mechanisms to detect fault conditions, such as
Classification of RRT air embolism or circuit occlusion.

In 1977, when Peter Kramer firstfi described con- Mechanism of Solute Removal
tinuous arteriovenous hemofi filtration (CAVH)
as a therapy for diuretic resistant flfluid overload Convection: when solute is cleared by ultrafiltra-

(5), the only other types of RRT were IHD and tion through a porous membrane, we say that
peritoneal dialysis (PD). Since then, the classifica-
fi the clearance of the solute is convective—carried
tion of RRT has expanded (6). See the glossary in by the bulk flow of plasma water. During CRRT,
Table 10.1. the process is called hemofiltration
fi .
Diffusion: when solute is cleared by diffusion
down a concentration gradient across a semi-
Duration or Timing of Therapy
permeable membrane, we say that the clearance
Continuous: CRRT aims to provide support 24 h/d, of the solute is diffusive. The process is called
but, in practice, is interrupted by factors such as hemodialysis.
patient transfer out of the ICU or circuit failure. Combinations: diafiltration
fi is the term applied
Intermittent: IHD requires approximately 3 hours when both convection and diffusion are operat-
per treatment. Patients with end-stage renal ing to remove solute.
failure (ESRF) may be maintained in the com- Adsorption: adsorption is the binding of sub-
munity with as few as three dialysis sessions per stances to the membrane under molecular
10. Renal Replacement Therapy 53

TABLE 10.1. RRT: A glossarya


Acronym Full Name Notes Seed reference
SCUF Slow continuous ultrafiltration AV or VV Silverstein 1974 (12)
CAVH Continuous arteriovenous hemofiltration Kramer 1977 (5)
CAVHD Continuous arteriovenous hemodialysis
CAVHDF Continuous arteriovenous hemodiafiltration
CVVH Continuous venovenous hemofiltration
CVVHD Continuous venovenous hemodialysis
CVVHDF Continuous venovenous hemodiafiltration
CHFD Continuous high flux dialysis AV or VV Ronco 1996 (13)
HVHF High volume hemofiltration AV or VV Cole 2001 (14)
CPF Continuous plasma filtration AV or VV Reeves 1999 (15)
CPFA Coupled plasma filtration adsorption AV or VV Ronco 2003 (16)
SLED Sustained low efficiency dialysis Marshall 2004 (17)
EDD Extended daily dialysis Kumar 2000 (18)
PDIRRT Prolonged daily intermittent RRT Naka 2004 (9)
a
AV, arteriovenous; VV, venovenous.

attraction. Adsorption is used specifi fically for There is little information regarding what spe-
toxin removal using activated charcoal car- cific
fi threshold plasma concentrations of potas-
tridges (10) and it is being tested as a means of sium, bicarbonate, urea, or creatinine should be
blood purifification in sepsis (11). used for the institution of RRT. In a retrospective
comparison of early versus late CRRT in trauma
associated ARF, Gettings et al. (23) found that
Intensity of Therapy patients in whom CRRT was commenced at a
mean blood urea nitrogen (BUN) of 42.6 mg/dL
For example, slow continuous ultrafiltration

(15.2 mmol/L) had a survival rate of 39% com-
(SCUF) is performed to simply remove excess
pared with 20% in patients in whom the
extracellular flfluid. High-volume hemofi filtration
mean BUN at commencement was 94.5 mg/dL
(HVHF) is used to intentionally accelerate the
(33.7 mmol/L).
clearance of target mediators, and high-fl
flux dialy-
sis (HFD) (dialysis performed with highly perme-
able membranes) is designed to accelerate the “Nonrenal” Indications
clearance of urea and larger molecules.
• Drug and toxin removal
• Sepsis and septic shock
• Inborn errors of metabolism
Indications for RRT • Congestive cardiac failure
• Cerebral edema
“Traditional” indications (19)
IHD can accelerate the elimination of small
• Diuretic resistant fluid overload
(<500 mw) unbound toxins with a low volume of
• Life-threatening hyperkalemia
distribution and minimal plasma protein binding
• Severe metabolic acidosis
(11), e.g., lithium, methanol, ethylene glycol, and
• Symptomatic uremia
salicylates. Continuous hemofiltration
fi has been
Kramer’s original description of CAVH involved used in lithium toxicity (24), with better hemody-
the treatment of diuretic resistant fl
fluid overload namic stability (25) but lower solute clearance
(5). During the intervening years, there has been than IHD.
controversy surrounding the use of diuretics in The use of extracorporeal blood purification

ARF (20), but they may be helpful in the fl fluid techniques in sepsis and septic shock is attractive
management of ARF before the institution of RRT but unproven. There were early observations of
(21, 22). improved cardiovascular and respiratory function
54 J.H. Reeves

in patients with severe sepsis after commence- of distribution of urea was cleared during the
ment of continuous hemofiltration
fi (26). This, session. Using a single compartment exponential
together with the identification
fi of inflflammatory washout model, we can predict that the final fi
mediators in filtrate
fi (27), led to efforts to increase concentration of the solute is approximately
infl
flammatory mediator removal during RRT. 37% of the starting concentration when the Kt/V
High-volume conventional hemofiltration
fi (14, 28), is 1.0.
large-pore hemofiltration
fi (29), and plasma filtra- In chronic renal failure, a minimum Kt/V of 1.2
tion with (30) or without (15) coupled adsorption should be delivered three times per week (36).
have been examined in small clinical trials. There There has been one randomized controlled trial
is currently no Level I evidence for the use of assessing dose of dialysis in ARF (7). Schiffl fl com-
extracorporeal blood purifi fication therapy in pared daily dialysis with alternate daily dialysis in
sepsis. 160 patients with ARF. The 28-day mortality using
End-stage cardiac failure is characterized by intention-to-treat analysis was 28% in the daily
progressive fl fluid retention, renal impairment, treated patients and 46% in the patients treated
neurohumoral stimulation, and diuretic resistance every other day. Multiple other outcomes were
(31). It was shown that patients with advanced improved in the daily dialysis group. Although this
cardiac failure can tolerate substantial fluid fl trial was controversial (37), it suggests that dialy-
removal during ultrafiltration
fi (12), with salutary sis every second day is insufficient
fi in critically ill
effects that persist beyond the time of fl fluid patients with ARF.
removal. Improved renal function, decreased heart Quantifi fication of clearance during CRRT can be
failure scores, lowered B natriuretic peptide levels, likened to the calculation of creatinine clearance,
decreased hospital length of stay, and fewer read- which is described by the formula UV/P. U is the
missions have all been observed in case-controlled urine concentration of the solute, V is the volume
studies (32). Most recently, a small randomized collected in a given time, and P is the plasma con-
controlled study showed that early ultrafiltration
fi centration. During CRRT, if we know the volume
results in increased weight loss at 24 h compared of efflfluent produced in a given time and the con-
with diuretics alone (33). Larger studies are war- centration of solute (e.g., urea) in both effluent

ranted for this indication. and plasma, we can calculate its clearance (38).
Cerebral edema can complicate IHD (34) and The result is a value in milliliters per minute. To
contribute to the clinical picture of disequilib- simplify the estimate further, let us assume that
rium. In patients with hepatic encephalopathy, the sieving coefficient
fi is 1.0, and that there is full
early studies compared the effects of IHD and concentration equilibrium between plasma water
CRRT (35). CRRT was associated with less decrease and dialysate. Then the effluent
fl concentration will
in mean arterial pressure, less increase in intracra- equal the plasma concentration, and clearance is
nial pressure, and less change in cerebral perfu- simply equal to the rate of production of effluent

sion pressure. from the hemofi filter.
Ronco, in 2000 (39), randomized 425 critically
ill patients with ARF to three different doses
Choosing the Dose and Mode of RRT (ultrafiltration
fi rates) of CRRT: 20, 35, or 45 mL/h/
kg. Fifteen-day survivals were 41%, 57%, and 58%,
Dialysis dose is a concept familiar to nephrologist respectively. This landmark study was one of the
in the setting of ESRF: Kt/V. first to formally adjust dose of CRRT based on

K is clearance (the volume of solute, usually patient weight. Importantly, it suggests that there
urea, cleared in a given time), t is duration of treat- is a threshold minimum level of clearance required
ment, and V is volume of distribution of the solute. for adequate CRRT of approximately 35 mL/h/kg.
Kt is the volume of plasma water cleared of solute Debate regarding the relative merits of CRRT
during the session, and Kt/V is Kt as a proportion and IHD has continued for nearly 30 years. From
of its volume of distribution. the outset (5), the attraction of CRRT was its sim-
For example, a Kt/V of 1.0 for urea means that plicity and cardiorespiratory stability compared
a total volume of plasma water equal to the volume with IHD. Now that CRRT is as complex as IHD
10. Renal Replacement Therapy 55

and potentially more expensive to perform for of an artifi ficial kidney, prognosis. Am J Med d 1955;
long periods, there is a paucity of good compara- 18:187.
tive studies that report outcomes such as mortal- 3. Mehta RL, Pascual MT, Soroko S, et al. Spectrum of
ity or recovery of renal function. In 2001, Mehta acute renal failure in the intensive care unit: the
PICARD experience. Kidney Intt 2004; 66(4):
randomized 166 critically ill patients to CRRT or
1613–1621.
IHD (40). The observed mortality was higher in
4. Meyer TW, Walther JL, Pagtalunan ME, et al. The
the group receiving CRRT, but this group had a clearance of protein-bound solutes by hemofiltra- fi
higher severity of illness. In 2002, Kellum pub- tion and hemodiafi filtration. Kidney Intt 2005; 68(2):
lished a meta-analysis of 13 trials, 3 randomized 867–877.
and 10 observational, comparing IHD and CRRT 5. Kramer P, Wigger W, Rieger J, et al. Arteriovenous
(41). Overall, there was no difference in mortality, haemofi filtration: A new and simple method for
but only six studies compared groups of equal treatment of over-hydrated patients resistant to
severity. In these six studies, the mortality was diuretics. Klin Wschr 1977; 55: 1121–1122.
lower in patients treated with CRRT. Since then, a 6. Ronco C, Bellomo R. Continuous renal replacement
well-designed prospective randomized controlled therapy: evolution in technology and current nomen-
clature. Kidney Int Suppl 1998; 66: S160–164.
trial has been published comparing CRRT with
7. Schifflfl H, Lang SM, Fischer R. Daily hemodialysis
IHD in 80 critically ill patients with ARF. There
and the outcome of acute renal failure. N Engl J Med
was no difference in survival or renal recovery 2002; 346(5): 305–310.
between the two groups. Although there was more 8. Marshall MR, Golper TA, Shaver MJ, Chatoth DK.
hemodynamic disturbance during IHD, this did Hybrid renal replacement modalities for the criti-
not translate to a survival benefifit for CRRT (42). cally ill. Contrib Nephrol 2001(132): 252–257.
9. Naka T, Baldwin I, Bellomo R, Fealy N, Wan L. Pro-
longed daily intermittent renal replacement therapy
Summary in ICU patients by ICU nurses and ICU physicians.
Int J Artif Organs 2004; 27(5): 380–387.
• RRT reduces the mortality of ARF from more 10. Zimmerman JL. Poisonings and overdoses in the
intensive care unit: general and specifi fic manage-
than 90% to approximately 50%.
ment issues. Crit Care Med 2003; 31(12): 2794–
• There is no proven difference in outcome
2801.
between intermittent and CRRTs, as long as a 11. Nalesso F. Plasma filtration adsorption dialysis
minimum dose of Kt/V of 6 to 8 per week is (PFAD): a new technology for blood purification. fi
achieved during hemodialysis or a clearance Int J Artif Organs 2005; 28(7): 731–738.
greater than 35 mL/kg/h is achieved during con- 12. Silverstein ME, Ford C, Lysaght MJ, Henderson LW.
tinuous hemofi filtration. Treatment of severe fl fluid overload by ultrafifiltration.
• New hybrid therapies (slow long extended daily New Eng J Med d 1974; 291(15): 747–751.
dialysis [SLEDD], extended daily dialysis [EDD], 13. Ronco C, Bellomo R, eds. Continuous high fl flux
and prolonged daily intermittent RRT [PDIRRT]) dialysis: an effificient renal replacement. Heidelberg:
may avoid the destabilization associated with Springer Verlag; 1996.
14. Cole L, Bellomo R, Journois D, et al. High-volume
IHD, and mitigate the cost and inconvenience of
haemofi filtration in human septic shock. Intensive
continuous hemofiltration.

Care Med 2001; 27(6): 978–986.
15. Reeves JH, Butt WW, Shann F, et al. Continuous plas-
References mafi filtration in sepsis syndrome. Plasmafi filtration in
Sepsis Study Group. Crit Care Med d 1999; 27(10):
1. Brivet FG, Kleinknecht DJ, Loirat P, Landais PJ. 2096–2104.
Acute renal failure in intensive care units-causes, 16. Ronco C, Brendolan A, d’Intini V, et al. Coupled
outcome, and prognostic factors of hospital mortal- plasma filtration
fi adsorption: rationale, technical
ity; a prospective, multicenter study. French Study development and early clinical experience. Blood
Group on Acute Renal Failure. Crit Care Med 1996; Purif 2003; 21(6): 409–416.
24(2): 192–198. 17. Marshall MR, Ma T, Galler D, et al. Sustained low-
2. Smith LH, Post RS, et al. Post traumatic renal insuf- effificiency daily diafifiltration (SLEDD-f) for critically
ficiency in military casualties. II. Management, use ill patients requiring renal replacement therapy:
56 J.H. Reeves

towards an adequate therapy. Nephrol Dial Trans- in septic shock. Crit Care Med 2002; 30(6): 1250–
plantt 2004; 19(4): 877–884. 1255.
18. Kumar VA, Craig M, Depner TA, Yeun JY. Extended 31. Ellison DH. Diuretic therapy and resistance in con-
daily dialysis: A new approach to renal replacement gestive heart failure. Cardiology 2001; 96(3-4): 132–
for acute renal failure in the intensive care unit. Am 143.
J Kidney Dis 2000; 36(2): 294–300. 32. Costanzo MR, Saltzberg M, O’Sullivan J, Sobotka P.
19. Palevsky PM. Renal replacement therapy I: indica- Early ultrafiltration
fi in patients with decompensated
tions and timing. Crit Care Clin 2005; 21(2): 347– heart failure and diuretic resistance. J Am Coll
356. Cardiol 2005; 46(11): 2047–2051.
20. Mehta RL, Pascual MT, Soroko S, Chertow GM. 33. Bart BA, Boyle A, Bank AJ, et al. Ultrafiltration

Diuretics, mortality, and nonrecovery of renal func- versus usual care for hospitalized patients with
tion in acute renal failure. JAMA 2002; 288(20): heart failure: the Relief for Acutely Fluid-
2547–2553. Overloaded Patients With Decompensated Conges-
21. Cantarovich F, Rangoonwala B, Lorenz H, et al. tive Heart Failure (RAPID-CHF) trial. J Am Coll
High-dose furosemide for established ARF: a pro- Cardiol 2005; 46(11): 2043–2046.
spective, randomized, double-blind, placebo- 34. Walters RJ, Fox NC, Crum WR, et al. Haemodialysis
controlled, multicenter trial. Am J Kidney Dis 2004; and cerebral oedema. Nephron 2001; 87(2): 143–147.
44(3): 402–409. 35. Davenport A, Will EJ, Davison AM, et al. Changes in
22. Uchino S, Doig GS, Bellomo R, et al. Diuretics and intracranial pressure during machine and continu-
mortality in acute renal failure. Crit Care Med d 2004; ous haemofi filtration. Int J Artif Organs 1989; 12(7):
32(8): 1669–1677. 439–444.
23. Gettings LG, Reynolds HN, Scalea T. Outcome in 36. Eknoyan G, Levin N. NKF-K/DOQI Clinical Practice
post-traumatic acute renal failure when continuous Guidelines: Update 2000. Foreword. Am J Kidney Dis
renal replacement therapy is applied early vs. late. 2001; 37(1 Suppl 1): S5–6.
Intensive Care Med d 1999; 258: 805–813. 37. Drazen JM, Ingelfinger
fi JR, Curfman GD. Removal of
24. van Bommel EF, Kalmeijer MD, Ponssen HH. Treat- expression of concern: Schiffl fl H, et al. Daily hemo-
ment of life-threatening lithium toxicity with high- dialysis and the outcome of acute renal failure. N
volume continuous venovenous hemofi filtration. Am Engl J Med 2002; 346: 305–310. N Engl J Med 2003;
J Nephrol 2000; 20(5): 408–411. 349(20): 1965.
25. Maggiore Q, Pizzarelli F, Dattolo P, et al. Cardio- 38. Reeves JH, Butt WW. A comparison of solute clear-
vascular stability during haemodialysis, haemo- ance during continuous hemofiltration,
fi hemodiafi fil-
filtration and haemodiafi filtration. Nephrol Dial tration, and hemodialysis using a polysulfone
Transplant 2000; 15 Suppl 1: 68–73. hemofi filter. ASAIO J 1995; 41(1): 100–104.
26. Gotloib L, Barzilay E, Shustak A, et al. Hemofiltra-
fi 39. Ronco C, Bellomo R, Homel P, et al. Effects of differ-
tion in septic ARDS. The artificial fi kidney as an ent doses in continuous veno-venous haemofiltration

artifi
ficial endocrine lung. Resuscitation 1986; 13(2): on outcomes of acute renal failure: a prospective
123–132. randomised trial. Lancett 2000; 356(9223): 26–30.
27. Tonnesen E, Hansen MB, Hohndorf K, et al. Cyto- 40. Mehta RL, McDonald B, Gabbai FB, et al. A random-
kines in plasma and ultrafi filtrate during continuous ized clinical trial of continuous versus intermittent
arteriovenous haemofi filtration. Anaesth Intensive dialysis for acute renal failure. Kidney Intt 2001;
Care 1993; 21(6): 752–758. 60(3): 1154–1163.
28. Cole L, Bellomo R, Hart G, et al. A phase II random- 41. Kellum JA, Angus DC, Johnson JP, et al. Continuous
ized, controlled trial of continuous hemofiltration
fi versus intermittent renal replacement therapy: a
in sepsis. Crit Care Med d 2002; 30(1): 100–106. meta-analysis. Intensive Care Med d 2002; 28(1):
29. Uchino S, Bellomo R, Goldsmith D, et al. Super high 29–37.
flux hemofi filtration: a new technique for cytokine 42. Augustine JJ, Sandy D, Seifert TH, Paganini EP. A
removal. Intensive Care Med d 2002; 28(5): 651–655. randomized controlled trial comparing intermit-
30. Ronco C, Brendolan A, Lonnemann G, et al. A pilot tent with continuous dialysis in patients with ARF.
study of coupled plasma filtration
fi with adsorption Am J Kidney Dis 2004; 44(6): 1000–1007.
11
Technical Aspects of Renal
Replacement Therapy
Sara Blakeley

Since its inception in 1977 (1), methods and equip- CRRT on the ICU nowadays is almost exclu-
ment used for the delivery of continuous renal sively venovenous in nature; in other words, blood
replacement therapy (CRRT) has undergone many is removed from a large vein and returned to a
changes. Intermittent hemodialysis (IHD) is per- large vein. The different modes of CRRT differ
formed on some intensive care units (ICU), but this mainly in their method of solute removal, and
chapter will concentrate on continuous therapies. most modern machines are capable of providing
the full range of modalities. Newer machines offer
greater ease in switching between modes.
Mode of CRRT There is much discussion regarding the rela-
tionship between dialysis dose delivered and sur-
There is a spectrum of treatment available and vival (6). A prospective randomized study of post
with choice comes debate; continuous versus dilution CVVH in critically ill patients found
intermittent, convective versus diffusive therapy improved survival with an ultrafiltration
fi dose of
(2, 3). Continuous therapies have been associated 35 ml/kg/hour (7). Ultrafiltration
fi rate is used as a
with better renal survival compared with IHD, dose surrogate. This finding was not repeated in a
although no compelling effect on overall mortality subsequent study (8) but a more recent study
has been seen (4). found did find an improvement in survival when
Diffusion is the movement of molecules from adding a dialysis dose to CVVH (9). It is unclear
an area of high concentration (blood) to one of whether this survival advantage was due to a
a lower concentration (dialysis fl fluid circulating higher dose of CRRT overall or due to the addition
in a counter current direction) across a semi- of a diffusive therapy to a convective one. These
permeable membrane. With convection a pressure findings are being investigated further in other

(transmembrane pressure) is applied across the studies, to clarify what is the optimal dose and
membrane, this drives water out and carries with means of delivery. In the mean time 35 ml/kg/hour
it dissolves solutes (solvent drag). The “waste” fl
fluid is often recommended as minimum that should be
produced is termed ultrafiltrate.
fi Both diffusive provided (10–12).
and convective therapies are good at removing Standard therapies in use are:
small molecular weight molecules (<5000 Da),
such as urea and creatinine, but “middle mole- Continuous venovenous hemofi filtration (CVVH),
cules” (10,000–50,000 Da) such as β2 microglobu- characterized by predominantly convective
lin and cytokines (5) may be better removed solute clearance (Figure 11.1A).
by convection. Currently, there is no definitive
fi Continuous venovenous hemodialysis (CVVHD),
evidence to suggest one particular mode over characterized by predominantly diffusive solute
another, and choice is often guided by local clearance but with some convection occurring
expertise. because of ultrafi
filtration.

57
58 S. Blakeley

Effluent pump

Ultrafiltrate and Blood flow from patient


used dialysate - ‘arterial’ side
(waste)
Blood pump

Replacement fluid pump

Replacement fluid
entering pre filter (pre
dilution)

Blood flow to patient


- ‘venous’ side

Haemofilter
Pressure monitor
Air detector
A

Anticoagulation syringe
Effluent pump

Ultrafiltrate and Blood flow from patient


used dialysate - ‘arterial’ side
(waste)
Blood pump

Replacement fluid pump


Dialysate fluid
(running in a
counter current Replacement fluid
entering post filter
direction to blood (post dilution)
flow through the
haemofilter)

Dialysate pump Blood flow to patient


- ‘venous’ side

Haemofilter
Pressure monitor
Air detector
B

FIGURE 11.1. A, CVVH with prefilter replacement fluid delivery (predilution). B, CVVHDF with postfilter replacement fluid delivery
(postdilution).
11. Technical Aspects of Renal Replacement Therapy 59

Continuous venovenous hemodiafiltration


fi Infusion rates of replacement flfluid and/or dial-
(CVVHDF), characterized by a mixture of ysate are set, with newer machines offering
diffusive and convective solute clearance increased fluid removal rates (0–10 L/h) to perform
(Figure 11.1B). high-volume hemofiltration
fi (HVHF). A desired
Slow continuous ultrafifiltration (SCUF) is removal net fl
fluid loss (0–1000 mL/h) is set by the operator
of water in response to a pressure gradient, but and constantly monitored by the machine. The
a degree of solute removal via convection will machine generally calculates the ultrafiltration

also occur. rate depending on the rate of replacement fl fluid
Therapeutic plasma exchange (TPE). Plasma is and fluid
fl removal set by the operator. A bag then
separated, removed, and replaced with either collects the effl
fluent (waste), which is composed of
albumin or fresh frozen plasma. ultrafi
filtrate and spent dialysate (if being used).
A series of safety mechanisms are in place to
prevent the inadvertent introduction of air to the
Hemofiltration Machines (Figure 11.2) patient and to detect blood leakage. Alarms warn
of pressure changes within the circuit:
All machines have a similar set up. Newer
machines differ in their degree of “user friendli- • Access pressure: Pressure in the “arterial” limb
ness,” their range of pump fl flow rates, and the removing blood from the patient is a negative
degree of monitoring. pressure reflecting
fl blood suction. It is deter-
A roller pump controls the flowfl of blood from mined by blood flow,fl patient’s blood pressure,
the patient through the fifiltration circuit and then and intravascular fluid status. Excessively
back to the patient. Blood flow
fl rate is set by the negative pressures (e.g., >300 mmHg) risk vascu-
operator but is limited by vascular access quality, lar injury and hemolysis and suggest occlusion
blood viscosity, patency of the hemofilter,
fi and car- somewhere in the access limb of the blood circuit,
diovascular stability of the patient. The maximal e.g., kinked line or blocked vascular access.
achievable blood flow
fl rate varies between differ- • Filter pressure: A rise in pressure indicates that
ent makes of machine (50–450 mL/min). the filter may be starting to clot.
• Return pressure: Pressure in the “venous” limb
returning blood to the patient is a positive pres-
sure reflflecting resistance to venous return. Low
pressures may indicate disconnection but can be
seen with changes in position. High pressures
are seen with occlusion to flow, e.g., kinking or
clotting, but also if the blood fl flow rate is too
high.

Replacement and Dialysis Fluid


With convective therapies, large volumes of fl fluid
(effluent)
fl are removed from the patient per hour.
To maximize solute removal, prevent the patient
from becoming hypovolemic, and replace wanted
electrolytes, a replacement fluid
fl is infused. With
diffusive therapies, dialysate fluid
fl d is run in a
counterclockwise direction through the fluid fl
compartment of the fi filter, against the blood flow.
This flfluid can be formulated in-house, or more
FIGURE 11.2. Two examples of hemofiltration machines. The commonly as commercially prepared bags of
Edwards’ Aquarius System (left)
t and Hospal Prisma (right).
t sterile fluid.
60 S. Blakeley

The fluid contains a buffer, either lactate or molecular weight less than 50,000 Daltons (Da),
bicarbonate, and electrolytes (sodium, chloride, i.e., smaller than albumin. Membranes are com-
magnesium, and calcium). Typically, replacement posed of two substances, cellulose (e.g., cupro-
fluids are potassium- and phosphate-free, and
fl phan) and synthetic fibers (e.g., polysulphone,
these may need to be replaced as clinically indi- polyamide, or polyacrylonitrile).
cated. Glucose is often not present.

Membrane Characteristics
Prefilter Versus Postfilter Infusion
Biocompatibility
Replacement fluid
fl is infused into the circuit either
before the filter (predilution) or after the filter Contact of blood with the fi
filter surface can lead to
(postdilution). Predilution lowers the hematocrit complement and leucocyte activation, triggering
of the blood passing through the filter,
fi potentially the coagulation cascade and infl flammatory
reducing anticoagulation needs and allowing pathways. More “biocompatible” indicates less
increased ultrafifiltration rates. This is at the expense complement/leucocyte activation. Activation of
of less-effective solute clearance, therefore, an infl
flammatory mediators has been suggested as
increased ultrafifiltration rate is needed to achieve one mechanism leading to ongoing renal injury,
similar solute clearances. Newer machines allow a and, therefore, delay or nonreturn of renal func-
combination of predilution and postdilution. tion (4). Kidneys that have already been injured
because of reduced renal perfusion lose their
ability to autoregulate pressure changes and the
Lactate Versus Bicarbonate Buffer kidney becomes very sensitive to even small
changes in renal perfusion. Early reports that bio-
Both lactate- and bicarbonate-buffered solutions
incompatible cellulose-based membranes led to a
have been shown to be effective in correcting
worse outcome have been debated, but, currently,
metabolic acidosis (11, 13). Bicarbonate is pre-
the evidence is not robust enough to defi finitively
ferred in patients with a preexisting lactic acidosis
recommend a synthetic membrane over cellulose
(e.g., septic shock) or with liver failure (11) because
or modifiedfi cellulose membrane (15). As it is,
these patients may be unable to metabolize an
most membranes used in CRRT are synthetic
exogenous lactate load normally, thus, worsening
because they have a greater degree of flflux.
the acidosis. Lactate levels are often seen to rise in
other patient groups who have a lactate buffer, but
the signifificance of this is unclear because hyper- Flux
lactatemia is not always associated with an acido-
This is a measure of ultrafifiltration capacity and is
sis. Bicarbonate is a more physiological buffer, but,
based on the membrane ultrafiltration
fi coeffi
ficient.
because it is unstable in solution, it needs to be
A filter with a high permeability coeffi ficient to
added just before use. Studies have compared out-
water will allow more ultrafi filtration (high flux)
comes of bicarbonate versus lactate buffers but
and, hence, more convective transport. Permeabil-
the evidence is inconclusive. Lactate intolerance
ity is a measure of the clearance of middle molec-
has been arbitrarily defi fined as a rise of greater
ular weight molecules and high permeability is
than 5 mmol/L during CRRT (14), and a change to
seen with high flflux membranes (synthetic mem-
a bicarbonate buffer should be considered.
branes). It should be noted that high permeability
does not always equate to high urea clearance
(efficiency).

Hemofilters
Structure Vascular Access
Thousands of hollow fibers (membrane) are
bundled together forming a hemofilter fi with a When initially developed, CRRT used an arterial
large surface area of 0.6 to 1.2 m2. Pores in the and a venous catheter (arteriovenous) (1). A wide-
membrane allow the passage of molecules with a bore (11.5–13.5 French) dual-lumen vascular dial-
11. Technical Aspects of Renal Replacement Therapy 61

ysis (venovenous) catheter is now generally used. through the circuit can also result in the formation
Mostly composed of polyurethane, they can have of platelet microthrombi, which can occlude the
an antibiotic/antimicrobial coating. filter. Loss of the filter through clotting can lead to

Blood is pumped from the patient (arterial ineffective dialysis and patient blood loss, as well
side) through proximal side holes into one lumen, as being a drain on resources, both nursing and
and is then returned though a port at the distal tip financial. Methods such as predilution and ensur-

of the second lumen (venous side). High blood ing adequate vascular access can be used, but some
flows without high pressures are ideal catheter
fl form of anticoagulation for the extracorporeal
design requirements, and the dual-lumen design circuit is often needed. Remember: circuit failure is
allows continuity and reduces recirculation. more often caused by inadequate vascular access
Vascular catheters differ in length, diameter of rather than inadequate anticoagulation.
lumen, and positioning of ports, and some have an
extra lumen added for drug infusions. Remember:
No Anticoagulation
always assume that EACH catheter limb contains
heparin and aspirate at least 5 mL of blood before In the setting of deranged clotting (e.g., interna-
using. Cuffed dialysis catheters are generally not tional normalized ratio [INR] >2, activated partial
used on the ICU, but may be indicated in stable thromboplastin time [aPTT] >60 s) and/or throm-
patients who are free of infection, and who require bocytopenia (e.g., platelet count <50,000) or a
ongoing renal replacement therapy (RRT) (Table high risk of bleeding, further anticoagulation is
11.1). often not necessary or carries the risk of bleeding.
With adequate access and predilution, it is possi-
ble to run the circuit without any anticoagulation
Positioning for an acceptable period of time and achieve good
A correctly positioned catheter will have a better solute clearance.
blood flow; good access is the key to good dialysis.
The tip of a jugular or subclavian catheter should
Unfractionated Heparin
extend to the superior vena cava and rest 1 to 2 cm
above the right atrium. Too short, and there is Unfractionated heparin (UFH) is the most com-
the possibility of recirculation, whereas a catheter monly used extracorporeal anticoagulant. The
that is too long risks atrial perforation. Femoral circuit is often primed with heparin (e.g., 1000–
catheters should be longer (>20 cm) to reach the 10,000 IU) because it is highly negatively charged
inferior vena cava, therefore, minimizing recircu- and is absorbed onto the plastic circuit. Depend-
lation and achieving better fl flow rates. ing on the risk of bleeding, a bolus dose (e.g., 10–
20 IU/kg) can be administered and a continuous
infusion started. A low-dose infusion (<5 IU/kg/h)
Site of Catheter aims NOT to prolong the aPTT. If clotting occurs,
Debate continues regarding the ideal site for place- a medium dose can be considered (5–10 IU/kg/h),
ment of dialysis catheters in terms of safety of aiming for mild prolongation of the aPTT (1–1.4
insertion and infection risk. Where long term times normal) (16). Prefilter
fi heparin can be neu-
dialysis is a possibility, there is concern that tralized with postfilter
fi protamine (e.g., 1000 IU/h
subclavian catheters may be associated with an to 10 mg/h), called regional heparinization. If
increased incidence of subclavian stenosis/throm- patients require formal heparinization for condi-
bosis, creating long-term problems for arteriove- tions such as a pulmonary embolus, this should be
nous fistula formation. continued, no extra “fi filter” heparin is needed.
The methods, site of sampling, and frequency
of anticoagulation monitoring vary depending
Anticoagulation on local protocols. Commonly used methods are
the activated coagulation time (ACT) and aPTT.
As blood flows
fl through the filter and fluid is However, it should be remembered that there is
removed, viscosity increases and there is a ten- not always a linear correlation between dose of
dency toward filter clotting. Passage of blood heparin or degree of anticoagulation and filter
fi life.
62 S. Blakeley

Low Molecular Weight Heparin Regional Citrate Anticoagulation


Compared with UFH, low molecular weight Infused citrate complexes with calcium and pre-
heparin (LMWH) is considered more effective in vents activation of the coagulation cascade and
reducing fibrin deposition on dialyzer membranes platelets. Post dialyser, calcium is reinfused.
and, thus, preventing circuit clotting, however, its Regional citrate anticoagulation (RCA) is an effec-
superiority to UFH has not been proven in trials. tive form of anticoagulation, and because only
There is a reduced incidence of heparin-induced the circuit is anticoagulated, it is safe to use in
thrombocytopenia syndrome (HITS) compared patients at risk of bleeding. However, its side
with UFH, but it is more costly. With more experi- effects and complex infusion protocol have limited
ence LMWH use has increased in popularity. Stan- its widespread use. Side effects include citrate
dard markers of anticoagulation are not reliable toxicity if citrate is not metabolized rapidly
and anti-Xa levels should be monitored with pro- or adequately (e.g., in liver failure), hypernatre-
longed use (target, 0.25–0.35 U/mL) (16). However, mia, hyper/hypocalcemia, and metabolic alkalosis
there is not always a correlation between anti-Xa (each citrate molecule is metabolized to three
levels and filter life. bicarbonates).

TABLE 11.1. Complications of RRTa


Access related
Complications during insertion Bleeding, local trauma
Infection Systemic or local
Catheter-related thrombosis
Patient immobility Particularly with femoral lines
Circuit related
Membrane bioincompatibility See text
Air embolism
Blood loss Caused by clotted filters (common) or disconnection (rare)
Fluid balance errors
Hemolysis

Dialysis related
Hypotension Hypovolemia secondary to total volume removal or speed of removal, i.e., not allowing body
compartments to equilibrate (commonest) t
High pump speeds may be enough to precipitate hypotension in unstable patients
Life-threatening anaphylactoid hypersensitivity reactions have been described with the use of certain
membranes (e.g., polyacrylonitrile membranes, such as AN69) with concurrent ACEI therapy
Activation of inflammatory and vasodilatory mediators (e.g., bradykinin) related to membrane
bioincompatibility
Anticoagulation-related complications Local or systemic bleeding
Related to specific type of anticoagulant: e.g., HITS (heparin) and hypocalcemia (RCA)
Electrolyte disturbances Including hypokalemia, hypophosphatemia, and hypoglycemia
Acid-base disturbances Metabolic acidosis related to lactate buffer in replacement fluid if unable to handle a large exogenous
lactate load (e.g., liver failure, septic shock)
Metabolic alkalosis related to RCA
Temperature disturbances A degree of cooling always occurs, this may lead to “normothermia” in febrile patients (i.e., masking a
pyrexia) or cause marked hypothermia
Vitamin and micronutrient depletion Water-soluble vitamins, trace minerals, certain hormones (e.g., glucocorticoids), amino acids
Inappropriate prescribing of drugs Generally leads to underdosing while a patient is on the filter
Further renal injury Systemic hypotension (see above) reducing already compromised renal perfusion
Because of release of inflammatory mediators triggered by blood coming into contact with the filter
and tubing
a
ACEI, angiotensin converting enzyme inhibitor.
11. Technical Aspects of Renal Replacement Therapy 63

Prostacyclin ery of acute renal failure. Cur Opin Crit Care. 2005;
11: 548–554.
Prostaglandin (PG)-I2 is a natural anticoagulant 5. Ricci Z, Ronco C, Bachetoni A, et al. Solute removal
that is a potent antiplatelet agent and has been during continuous renal replacement therapy in
shown to reduce platelet microthrombi during critically ill patients; convection versus diffusion.
dialysis. It is often used in patients with a high risk Crit Care. 2006; 10: R67–R74.
of bleeding, but because it is a potent arterial 6. Clark WR, Turk JE, Kraus MA, Gao D. Dose deter-
vasodilator, some patients develop symptomatic minants in continuous renal replacement therapy.
hypotension. It has been used on its own and in Artif Organs. 2003; 27: 815–820.
7. Ronco C, Bellomo R, Homal P, et al. Effects of dif-
combination with low-dose heparin.
ferent dose in continuous veno-venous haemofiltra-fi
tion on outcomes of acute renal failure: a prospective
randomised trial. Lancet. 2000; 356: 26–30.
Other Anticoagulants 8. Bouman C, et al. Effects of early high-volume con-
Heparinoids (e.g., danaparoid) have minimal tinuous Venovenous hemofiltratin
fi of survival and
effects on platelets and can be used in HITS (but recovery of renal function in intensive care patients
remembering the potential cross reactivity in with acute renal failure: a prospective randomized
5–10% of patients). However, standard markers of trial. Crit Care Med. 2000; 30: 2205–2211.
9. Saudan P, et al. Adding a dialysis dose to continuous
anticoagulation are not reliable and its effect is
hemofi filtration increases survival in patients with
prolonged in renal failure. Factor Xa inhibitors acute renal failure. Kidney Int. 2006; 70: 1312–
(e.g., fondaparinux) and direct thrombin inhibi- 1317.
tors (e.g., recombinant hirudin) can be safely used 10. Cariou A, Vinsonneau C, Dhainaut JF. Adjunctive
in HITS, but, to date, have limited use in CRRT. therapies in sepsis: an evidence-based review. Crit
Care Med. 2004; 32: S562–S570.
11. www.adqi.net
Comment 12. Ronco C. Renal replacement therapy for acute
kidney injury: let’s follow the evidence. Int J Artif
A recent systematic review (16) found that there
Organs. 2007; 30: 89–94.
was no conclusive evidence to suggest one strat- 13. Naka T, Bellomo R. Bench-to-bedside review: treat-
egy over another, but the chosen method should ing acid-base abnormalities in the intensive care
take into account patient characteristics and local unit–the role of renal replacement therapy. Crit
facilities. Heparin (UFN and LMWH) has the Care. 2004; 8: 108–114.
greatest evidence and experience behind it, but 14. Hilton PJ, Taylor J, Forni LG, Treacher DF. Bicarbo-
RCA is increasing in popularity and ease of use. nate-based haemofiltration
fi in the management of
acute renal failure with lactic acidosis. QJM. 1998; 4:
279–283.
References 15. Teehan GS, Liangos O, Lau J, et al. Dialysis mem-
1. Kramer P, Wigger W, Rieger J, et al. Arteriovenous brane and modality in acute renal failure: under-
haemofi filtration: a new and simple method for treat- standing discordant meta-analyses. Sem Dialysis.
ment of over-hydrated patients resistant to diuret- 2003; 16: 356–360.
ics. Klin Wochenschr. 1977; 55: 1121–1122. 16. Oudemans-van Straaten HM, Wester JPJ, de Pont
2. Palevsky PM. Dialysis Modality and Dosing Strat- ACJM, Schetz MRC. Anticoagulation strategies in
egy in Acute Renal Failure. Sem Dialysis. 2006; 19: continuous renal replacement therapy: can the
165–170. choice be evidence based? Intensive Care Med. 2006;
3. Van Biesen W, Vanholder R, Lameire N. Dialysis 32: 188–202.
strategies in critically ill acute renal failure patients.
Curr Opin Crit Care. 2003; 9: 491–495. Suggested Reading
4. Palvesky PM, Baldwin I, Davenport A, et al. Renal
replacement therapy and the kidney: minimising Bellomo R, Baldwin I, Ronco C, Golper T. Atlas of Hemo-
the impact of renal replacement therapy on recov- fi
filtration. WB Saunders. 2002.
12
End-Stage Renal Disease
Emile Mohammed

There are now approximately one million people Hemodialysis


on renal replacement therapy worldwide. In the
current era of chronic noncommunicable disease, Many hemodialysis (HD) techniques have been
this number is set to double within the next developed, particularly in the ICU setting, ranging
decade. Patients with end-stage renal disease from conventional HD, high-fluxfl HD, hemodiafi fil-
(ESRD) carry a significantly
fi higher cardiovascular tration, and hemofi filtration. These techniques
morbidity and mortality compared with the simply use varying degrees of convection or
general population. This is because of the “uremic” diffusion. For example, hemofiltration
fi is a convec-
cardiovascular factors (Table 12.1). The result is tive treatment with good clearance of mid-size
that there will be a growing number of ESRD molecules but with poor small molecule clear-
patients being managed within the intensive care ance. The converse holds for conventional inter-
unit (ICU) setting. mittent HD.

Hemodialysis and Peritoneal Dialysis Peritoneal Dialysis


Mechanisms of Dialysis
The peritoneum acts as a natural semipermeable
There are two basic principles of dialysis that membrane. Dissolved waste products and water
allow the body’s homeostasis to be achieved in the pass from the blood, via the peritoneal capillaries,
absence of a natural kidney. They are as follows: through the mesothelial cells and interstitium to
the peritoneal dialysis (PD) fl fluid (PDF). This
• Convection, in which there is movement (in
process is referred to as ultrafi filtration (Figure
large volumes) of solvent, which drags dissolved
12.2). Water-soluble waste products pass down a
solute across a membrane with a hydrostatic
concentration gradient that is generated by an
pressure gradient.
osmotic gradient. This, in turn, is created by
• Diffusion, in which there is passive movement
glucose or glucose polymers added to the PDF.
of solute from a high- to a low-concentration
PD regimens are all based on repetitions of a
gradient across a membrane (Figure 12.1). Dif-
basic cycle, which comprises inflow
fl of PDF, a dwell
fusion depends not only on the transmembrane
time of the PDF within the peritoneal cavity, and
gradient but the membrane characteristics (e.g.,
then drainage. The various types of PD are all
pore size).
based on this principle. They include continuous
Diffusion is more effective in clearing small ambulatory PD (CAPD), automated PD (APD),
molecules and convection improves mid-size mol- tidal PD, and intermittent PD. Typical regimes are
ecule clearance. illustrated in Figure 12.3.

64
12. End-Stage Renal Disease 65

TABLE 12.1. Cardiovascular risk factors in the uremic patient


Traditional coronary risk factors Uremic-related cardiovascular
(The Framingham Study) risk factors
Hypertension Increased extracellular fluid
volume
High levels of low-density Calcification and high
lipoprotein calcium/phosphate product
Low levels of high-density Parathyroid hormone
lipoprotein
Smoking Anemia
Diabetes Oxidant stress
Older age Malnutrition
Male sex Pulse pressure
White race Triglycerides
Physical inactivity Lipoprotein remnants
Menopause Lipoprotein A
Left ventricular hypertrophy Homocysteine
Inflammation (C-reactive protein)
Sleep disorders

Clinical Parameters of Peritoneal Dialysis in Mexico (ADEMEX) study


(2) again reveals the same controversy of defining

Much debate surrounds the “optimal” dialysis the optimal dialysis dose in PD patients. In this
dose, although a minimal dialysis dose has been study, there was a neutral effect on patient survival
universally accepted. Within the context of thrice- between a control group on conventional CAPD
weekly HD, there seems to be no added benefit fi of compared with a study group on a modified fi pre-
high-dose dialysis compared with the conven- scription, which achieved increased small solute
tional dose of dialysis (the HEMO study) (1). clearance, measured by peritoneal creatinine
Dialysis adequacy is measured by urea kinetic clearance and peritoneal Kt/V.
modeling (UKM) using the urea reduction ratio, There are no “fi fixed targets” that determine
or Kt/V, where K is the dialyzer urea clearance, t dialysis adequacy in the ICU setting. Dialysis
is the duration of dialysis, and V is the urea distri- dose and duration must be determined by
bution volume. In a well-nourished stable HD balancing the clinical condition of the patient,
patient, a Kt/V of 0.8 to 1.0 is the minimum accept- while achieving as normal a physiological state as
able threshold per dialysis session. The Adequacy possible.

Blood flow (250-500ml/min)

Sodium Calcium, bicarbonate

Semi
em
mi-pe
-pe
-perme
perm
rmeab
rmeablle
memb
em
mbbran
ranee

Urea, creatinine Water Potassium


FIGURE 12.1. Diagrammatic representa-
tion of blood purification within the Dialysate flow (400-800ml/min)
dialyzer.
66 E. Mohammed

Interstitium Mesothelium Peritoneal cavity FIGURE 12.2. Ultrafiltration in PD.

Soluble waste products and water

Glucose

Capillary

Typcal CAPD regime

3
“Bag in”
2.5
PDF volume (litres)

1.5

0.5

0
Bag drained out Time (during day)

Typical APD regime

2.5

“Bag in”
PDF volume (litres)

1.5

0.5

0
wet day overnight exchanges FIGURE 12.3. PD regimes.
12. End-Stage Renal Disease 67

The following clinical parameters act as guide- • Anaphylaxis. Anaphylaxis can occur by comple-
lines to achieve this: ment activation with the use of a bioincompati-
ble membrane and normally occurs within the
• Target weight and blood pressure control. Target first 20 minutes of treatment.
weight is defifined as the patient’s weight in which • Catheter related-sepsis. Catheter related-sepsis
all the fluid compartments are physiologically requires aggressive antibiotic treatment and
normal. Excess weight (which will be essentially catheter removal. If temporary catheters are
salt and water) results in hypertension. The being used, once weekly catheter changes are
target weight is achieved by gradual weight recommended.
reduction on successive dialyses until the • Pyrogenic reactions. Uncommon if ultrapure
patient is free from both pulmonary and peri- water is used.
pheral edema, but, below which, hypotension • Dialysis equilibrium syndrome. Rare in estab-
occurs. lished dialysis patients. It can occur from overag-
• Acid-base balance. Dialysis must be performed gressive dialysis causing a rapid reduction in
frequently and long enough to maintain normal serum osmolality and resulting in cerebral
acid-base balance. edema.
• Bone biochemistry. Along with vitamin D sup- • Modern fail-safe machines minimize other com-
plementation, serum calcium and phosphate plications such as air embolism and accidental
levels should be maintained within normal circuit disconnection.
limits.
• Nutritional state. It is important to remember
that a high proportion of ESRD patients within Peritoneal Dialysis
the ICU will have a low serum albumin, low Although PD is a technically safe procedure, there
body mass index, an infl flammatory and/or may be clinical reasons to convert to temporary
hypercatabolic state, and a low dietary intake. It HD.
is, therefore, necessary to obtain dietary advice, These are as follows:
treat correctable factors, give dietary supple-
ments, and have a low threshold for nasogastric • Abdominal surgery
(NG), percutaneous endoscopic gastrostomy • Diaphragmatic fluid leak resulting in effusions
(PEG), or even parenteral nutrition, if indicated. • Respiratory compromise from splinting of dia-
Dialysis prescriptions must accommodate these phragm by PDF
nutritional requirements. • Severe hypoalbuminemic state
• Peritonitis or catheter-related sepsis
A good starting point for prescribing dialysis • Inadequate ultrafiltration
fi in the context of
would be to continue the patient’s regular dialysis aggressive flfluid management and/or hypercata-
regime and adjust the dose of dialysis, in conjunc- bolic state of the patient
tion with the nephrologists, to achieve the above
parameters.
Renal Transplantation
Dialysis-Related Complications Renal transplantation represents the best mode of
therapy for ESRD patients, both in cost effective-
Hemodialysis ness and quality of life (3). There have been many
• Hypotension. Hypotension can be minimized improvements in renal transplantation, such as the
with an accurate assessment of target weight, refi
finement of immunosuppression regimens, and
judicious use of antihypertensive medications, patient-donor selection and work-up as well as
sodium restriction, increasing treatment dura- their compatibilities. The major challenge facing
tion, and careful choice of dialysis modality, e.g., transplantation is that its demand far outstrips the
hemodiafiltration
fi in the cardiovascularly unsta- availability. Every effort should be made to increase
ble patient. the number of donors. In parallel to this, there is
68 E. Mohammed

much research in the development of stem cell allografts. Such sensitization can cause severe
transplantation and xenotransplantation. hyperacute rejection.

Evaluation, Selection, and Evaluation and Selection of Donors


Preparation of the Potential There are two sources of donors:
Transplant Recipient
Cadaveric: Cadaveric kidneys may be either from
General Evaluation patients with brainstem death and a maintained
cardiac output or from nonheart-beating
There are few absolute contraindications to renal donors. Donors with sepsis, malignancy, infec-
transplantation. These are uncontrolled cancer, tion with hepatitis B, hepatitis C, HIV, or tuber-
HIV positivity, active systemic infections, and/or culosis, or irreversible renal failure are not
any condition with a life expectancy of shorter considered for donation.
than 2 years. Conditions increasing the risk of Live: The use of kidneys from living donors is
posttransplant morbidity and mortality include recommended for renal transplantation when-
long duration of dialysis, previous incidence of ever possible, in light of the growing body of
recurrent infections, cardiovascular disease, and evidence of favorable outcomes after transplan-
gastrointestinal complications. Such patients tation. Before being selected as a living donor,
require a particularly careful work-up and aggres- thorough counseling, medical, physical, and
sive management of risk factors (e.g., hyper- psychological evaluation is performed. Outcome
tension, obesity, and vascular disease) before studies have revealed lower mortality rates in
transplantation. living donors compared with the general popu-
lation. This is probably caused in part by patient
selection and the fact that this group of patients
Psychological Evaluation
receive long-term medical follow-up.
The use of psychiatric screening is not universally
adopted but may be useful in assessing compli-
ance with immunosuppressive regimes. Poor Immunosuppression
compliance signifificantly worsens renal allograft
outcomes. The immunosuppression regime is tailored to
each patient in an effort to minimize rejection
Recurrent Renal Disease as well as side effects. The following is a brief
summary of drugs used, usually in combination.
It is important to ascertain the underlying cause The commonest regime is triple therapy, e.g.,
of renal failure because some diseases recur in the cyclosporin, azathioprine, and prednisolone.
transplanted kidney, most notably, focal and seg- Corticosteroids have broad but potent immuno-
mental glomerulosclerosis. suppressive actions. They are used in high doses
in induction therapy as well as for episodes of
acute rejection. They are tapered to small mainte-
Immunological nance doses or stopped completely over time.
ABO blood group must be compatible. HLA typing Because of the broad actions of corticosteroids,
of donors and recipients allows assessment of there are a large range of side effects.
compatibility. HLA DR is more important than Cyclosporin is a calcineurin inhibitor. Although
HLA B, which is more important than HLA A. there is little myelotoxicity, cyclosporin is nephro-
A lymphocyte cross-match is also performed: the toxic and does contribute to chronic allograft
recipient is screened for preexisting antibodies to rejection. It is an important agent, because its
donor lymphocytes, which arise in response to introduction improved 1-year graft survival by 15
previous blood transfusions, pregnancies, or renal to 20%.
12. End-Stage Renal Disease 69

Tacrolimus is also a potent calcineurin inhibitor. Early


Its side effect profi
file is similar to cyclosporin but
seems to be more diabetogenic, particularly in the • Acute rejection should be suspected in patients
black population. with established graft function who then experi-
Azathioprine has been widely used for trans- ence a rise in serum creatinine. A biopsy is
plantation and it continues to be an integral part usually required to confirm
fi the clinical diagnosis
of many immunosuppression regimens. It inhibits and the treatment involves high-dose steroids,
purine metabolism and, therefore, cellular prolif- usually with some modification
fi of the immuno-
eration. There is a significant
fi side-effect profi
file, suppression regime. Antibodies are considered
most notably its myelosuppressive effect, which is in steroid-refractory acute rejection.
worsened by concomitant use of other drugs such • Infectious complications tend to vary with time
as allopurinol. after transplantation. Within the fi first month
Mycophenolate mofetill acts similarly to azathio- after transplantation, most infections are sur-
prine but is more specifi fic for lymphocytes. gically related, such as atelectasis and wound
Myelosuppression must also be monitored and infection. From 1 to 6 months after transplanta-
mycophenolate mofetil has more gastrointestinal tion, opportunistic infections can emerge, such
side effects than azathioprine. as Pneumocystis carinii and Aspergillus fumiga-
Daclizumab and basiliximab are anti-CD25 tus. Clinical infection caused by the effects of
antibodies. They target activated T cells only and modulating viruses, cytomegalovirus (CMV)
are used as an induction agent to prevent early and Epstein-Barr virus (EBV) are serious com-
rejection. plications for which there should be surveillance
Polyclonal antibodies to T-cells, ALG and ATG, as with prophylaxis administered where appropri-
well as monoclonal antibody to T cells, OKT3, are ate. PTLD in the early period tends to be an
used to treat refractory acute rejection and some- EBV-related malignancy.
times used as an induction agent to prevent rejec- • Mechanical complications, such as arterial and
tion. These drugs are used cautiously because their ureteric stenoses and lymphoceles exerting local
administration is associated with cytokine release pressure, sometimes evolve as a cause of deteri-
syndrome and pulmonary edema. Other side orating graft function.
effects include subsequent infection and posttrans-
plantation lymphoproliferative disease (PTLD).
Late
• Chronic rejection occurs secondary to a combi-
Complications of Transplantation nation of immunological and nonimmunolo-
gical factors. The result is an irreversible
Immediate
progressive decline in graft function and is often
• Acute tubular necrosis is the commonest cause associated with proteinuria.
of early graft function and is more likely to • Recurrence of original disease may result in graft
occur with prolonged ischemic times and asys- failure. There is a particularly high rate of recur-
tolic, hypotensive, or elderly donors. rence of focal and segmental glomerulosclerosis
• Surgical complications are now the commonest but it is diffi
ficult to predict and, therefore, is not
cause of early graft loss. These include renal vein a contraindication to transplantation.
and arterial thrombosis and urinary leaks. These • Cardiovascular disease is the main cause of
complications are surveyed for with Doppler death in the transplant population and the rate
ultrasound and isotope scanning. Occasionally of cardiovascular disease is higher than in the
renal angiography or surgical reexploration is general population. This is because of the addi-
required in the anuric kidney. tional risk factors of the ESRD population (Table
• Hyperacute rejection is now very uncommon, 12.1) as well as the adverse effects of the immu-
with the more meticulous screening for the nosuppressive agents.
presence of preexisting antibodies. The only • Malignancy, in particular, skin cancer, is three
available treatment is graft nephrectomy. times more common than the general population.
70 E. Mohammed

Other cancers that should be screened for include membrane flux in maintenance hemodialysis. N Engl
renal, cervical, and vaginal cancers. J Med 2002; 347(25): 2010–2019.
2. Paniagua R, Amato D, Vonesh E, et al. Health-related
quality of life predicts outcomes but is not affected
by peritoneal clearance: The ADEMEX trial. Kidney
Summary Intt 2005; 67(3): 1093–1104.
3. The EBPG Expert Group on Renal Transplantation.
ESRD and its complications are becoming more European Best Practice Guidelines for Renal Trans-
commonplace, particularly in the ICU environ- plantation (Part 1). Nephrol Dial Transplantt 2000;
ment. The challenges associated with this group 15(Supp 7): 1–85.
of patients also continue to rise and requires a
multidisciplinary approach, which includes the
intensivist and the nephrologist. Suggested Reading
Davison AM, Cameron S, Grunfeld J-P, et al. Oxford
References Textbook of Clinical Nephrology. 3rd edition. Oxford:
Oxford University Press. 2005.
1. Eknoyan G, Beck GJ, Cheung AK, et al. Hemodialysis Levy J, Brown E, Morgan J. Oxford Handbook of Dialy-
(HEMO) Study Group. Effect of dialysis dose and sis. Oxford: Oxford University Press. 2001.
13
Clinical Hyperkalemia and Hypokalemia
Harn-Yih Ong

Hyperkalemia excreting 90 to 95% of the daily potassium load.


Decreased renal excretion of potassium is, there-
Hyperkalemia is defined
fi as a plasma potassium fore, the most important cause of hyperkalemia.
concentration greater than 5.0 mmol/L and refers Hyperkalemia may follow failure of glomerular
to an excess concentration of potassium ions in filtration or tubular secretion of potassium.

the extracellular fl
fluid (ECF) compartment.

Causes of Hyperkalemia Clinical Features


(Tables 13.1 and 13.2) Hyperkalemia is often asymptomatic, but severe
hyperkalemia (potassium >6.5 mmol/L) may pre-
Because of the ability of the kidneys to excrete a sent with neuromuscular disturbances such as
large amount of potassium, hyperkalemia caused distal paresthesia, generalized muscle weakness,
by accelerated exogenous intake usually indicates an ascending flaccid
fl paralysis, or ventilatory
the presence of a subtle or overt defect in renal failure. Sudden cardiac death may occur when
potassium handling or an altered transcellular plasma potassium concentration exceeds 7.0 to
distribution. 7.5 mmol/L, and this may be the earliest and only
manifestation. Acute hyperkalemia is much less
Compartmental Shift well tolerated than chronic hyperkalemia.

Potassium is the principal intracellular cation, and


maintenance of the normal distribution of potas- Electrocardiogram Changes
sium between the intracellular and extracellular
compartments relies on several regulatory mecha- Some patients may show a gradual progression
nisms. Under normal conditions, ingested potas- of electrocardiogram (ECG) findings
fi but may
sium is absorbed into the portal circulation progress rapidly without warning. Cardiac arrest
and rapidly shifted into cells by Na+/K+-ATPase caused by asystole, ventricular tachycardia, or ven-
under the influence
fl of insulin and circulating β- tricular fi
fibrillation may occur at any point along
adrenergic catecholamines. When these mecha- this progression. Hyperkalemia can be life threat-
nisms are perturbed, hyperkalemia may occur. ening even if the ECG is normal. Approximately
50% of patients with potassium levels exceeding
6.0 mmol/L have a normal ECG.
Decreased Renal Excretion
The renal system maintains external potassium • K+, 5 to 6 mmol/L: peaked T waves and short-
balance in the long term and is responsible for ened QT interval

71
72 H.-Y. Ong

TABLE 13.1. Extrarenal causes of hyperkalemia


Increased intake
Exogenous sources Potassium supplements (especially with advanced age, diabetes mellitus, underlying renal
impairment, and/or the concurrent use of potassium-sparing diuretics)
Stored packed red blood cells. Cardiac tolerance to hyperkalemia is decreased by hypocalcemia
because of the presence of the anticoagulant, citrate
Potassium penicillin G
Salt substitutes
Endogenous sources (tissue breakdown) Rhabdomyolysis. Each kilogram of lean muscle mass contains more than 100 mmol of potassium
Hemolysis
Tumor lysis syndrome
Reperfusion syndrome. Ischemia results in acidosis in the affected area and an outflow of
intracellular potassium, this potassium is “washed out” when the region is reperfused
Catabolic states
Compartmental shift
Inhibition of Na+/K+-ATPase Insulin resistance/deficiency
β2-adrenergic catecholamine deficiency or resistance; e.g., β-blockers
Drugs and toxins; e.g., cardiac glycosides
Altered transcellular electrochemical Inorganic metabolic acidosis
K+ gradient ECF hypertonicity or hyperosmolar states; e.g., hyperglycemia, hypertonic solutions such as mannitol,
hypertonic saline
Increased conductance of K+ channels Drugs; e.g., depolarizing muscle relaxants (succinylcholine)
Familial hyperkalemic periodic paralysis

• K+, 6 to 7 mmol/L: prolonged PR interval, AV Pseudohyperkalemia


dissociation, flattening
fl and loss of P wave, and
widening of QRS complex Pseudohyperkalemia is caused by the release of
• K+, greater than 7 to 8 mmol/L: sine wave pattern, potassium from damaged cells in vitro (during
ventricular fibrillation, and asystole or after the removal of the blood sample from

TABLE 13.2. Renal causes of hyperkalemia


Inadequate sodium delivery to the cortical collecting duct
Intravascular volume depletion Acute circulatory failure, e.g., vasodilatory or septic shock
Severe cardiac failure
Hepatic cirrhosis with ascites
Renal failure Acute (oliguric) and chronic renal failure
Secondary hypoaldosteronism
Low renin Suppressed renin secretion, e.g., cyclosporin, tacrolimus
Distal (type IV) renal tubular acidosis, e.g., associated with diabetic nephropathy, tubular interstitial nephritis,
inhibitors of prostaglandin production
Normal/high renin Impaired aldosterone production, e.g., heparin
Impaired angiotensin II production/action, e.g., angiotensin-converting enzyme inhibitors, angiotensin II
receptor blockers
Normal/high renin, low cortisol Primary adrenal insufficiency, e.g., Addison’s disease (autoimmune)
Impaired adrenal enzyme biosynthesis, e.g., azole antifungals (ketoconazole), congenital adrenal enzyme
defects (rare)
Primary hypoaldosteronism (rare)
Tubular hyperkalemia without aldosterone deficiency (aldosterone resistance)
Renal distal tubular potassium Aldosterone antagonists, e.g., spironolactone
secretory defect (acquired) Luminal sodium channel blockade, e.g., amiloride, cimetidine, trimethoprim, pentamidine
Inhibition of basolateral Na+/K+-ATPase, e.g., cyclosporine
Acute or chronic tubulointerstitial nephritis, e.g., diabetic nephropathy, obstructive uropathy, postrenal
transplant, lupus nephritis, sickle cell nephropathy, amyloidosis
Pseudohypoaldosteronism Type I and II. Rare
13. Clinical Hyperkalemia and Hypokalemia 73

the patient) and can produce a 1 to 2 mmol/L arti- hyperglycemia is effective and rapid in onset of
factual increase in potassium levels. The major action. For example, 10 U intravenous insulin
causes are: can be expected to lower the plasma potassium
concentration by 0.5 to 1.5 mmol/L within 15
• Contamination by drip site or laboratory error
minutes, lasting 2 to 4 hours.
• Hemolysis caused by mechanical trauma during
• Sodium bicarbonate: Increasing the pH of the
venipuncture
ECF compartment with sodium bicarbonate
• Thrombocytosis (platelet count >900 × 1010
(e.g., 100 mmol over 1–2 hours) will shift potas-
cells/L)
sium into cells as an acidosis is corrected. Its
• Leukocytosis (white cell count >70 × 109 cells/L)
effectiveness is debated, but it is still recom-
or mononucleosis (“leaky red blood cells”)
mended when hyperkalemia is associated with
• Familial pseudohyperkalemia or other uncom-
severe inorganic metabolic acidosis. Ensur-
mon genetic syndromes
ing effective diuretic therapy fi
first lessens the
likelihood of developing volume overload as a
complication.
Management • Salbutamol: Meter-dose inhalers, nebulized, or
intravenous salbutamol seem equally effective
Severe hyperkalemia (plasma potassium
in reducing potassium levels.
>6.5 mmol/L or with ECG changes) is a medical
emergency and therapy should begin
immediately. Removal of Excess Potassium
Treatments that shift potassium into the cells have
Stabilization of Cardiac Membranes: no effect on total body potassium. Excess total
Intravenous Calcium body potassium may be removed by gastrointes-
tinal elimination, renal excretion, or renal replace-
Although no randomized studies exist to support ment therapy.
the use of calcium salts administered intrave-
nously (e.g., 10 mL 10% calcium gluconate over • Renal excretion of potassium may be enhanced
2–3 min into a large vein), it is still recommended with the use of diuretics, especially loop diuret-
as first-line therapy in the presence of ECG changes ics, which increase the delivery of sodium and
or arrhythmia. The protective effect of calcium is the urine flow rate to the cortical collecting duct,
evident within minutes and lasts for 30 to 60 increasing tubular secretion of potassium. If the
minutes. Caution should be used in patients who patient is volume depleted, hydration with saline
take digoxin because calcium has been reported can be administered with the diuretic.
to worsen the myocardial effects of digoxin toxic- • Although widely used clinically in the treatment
ity. An alternative is to consider using magnesium of hyperkalemia, cation exchange resins do not
instead of calcium to stabilize the myocardium. seem to be effective at 4 hours and should not
Calcium may reduce the immediate risk of cardiac be relied on for rapid effects.
arrest, but represents a temporizing measure only. • Hemodialysis or continuous renal replacement
Plasma potassium concentration is unaltered. therapies are the treatments of last resort, with
the exception of patients already receiving these
therapies.
Transcellular Redistribution
• Insulin and dextrose: Administration of insulin
always causes a transient reduction in plasma Hypokalemia
potassium because the activated insulin recep-
tor stimulates Na+/K+-ATPase, driving cellular Hypokalemia is usually defined
fi as a plasma potas-
uptake of potassium. The use of insulin and sium concentration of less than 3.5 mmol/L, and
glucose (e.g., 50 mL of 50% glucose to prevent refers to a deficit
fi of potassium ions in the ECF
hypoglycemia) for the emergency treatment of compartment.
74 H.-Y. Ong

Causes of Hypokalemia (Table 13.3) Familial Hypokalemic Periodic Paralysis

Hypokalemia indicates a disruption of normal Familial Hypokalemic Periodic Paralysis is an


potassium homeostasis and is almost always uncommon inherited (autosomal dominant) dis-
caused by potassium depletion caused by increased order characterized by recurrent episodes of
losses, either through the kidneys or gut. In either muscle paralysis that subside spontaneously after
case, drugs are the most common cause. Less fre- 6 to 24 hours. The primary defect is the gene encod-
quently, hypokalemia occurs because of an acute ing the skeletal muscle dihydropyridine receptor, a
shift of potassium from extracellular to intracel- voltage-gated calcium channel. Acute attacks are
lular fluid (ICF) compartments (compartmental precipitated by a reduction in plasma potassium,
shift). For any given cause, the magnitude of the such as after carbohydrate-rich meals (increased
change is relatively small (usually <1.0 mmol/L), insulin) or rest after exertion (increased catechol-
but a combination of factors may lead to a signifi-fi amines). Administration of potassium is lifesaving
cant fall in plasma potassium. and should be administered to treat acute attacks.

TABLE 13.3. Causes of hypokalemia


Inadequate intake of potassium (rare)
Compartmental shift
Stimulation of Na+/K+-ATPase-mediated Endogenous or exogenous insulin
cellular potassium uptake
β2-adrenergic receptors activation or indirect β2 sympathomimetics (e.g., salbutamol,
dobutamine)
Increased endogenous adrenaline release (e.g., delirium tremens, hypoglycemia, coronary
ischemia, after cardiopulmonary resuscitation)
Phosphodiesterase inhibition (e.g., xanthines: milrinone, theophylline, and caffeine)
Thyrotoxic hypokalemic periodic paralysis
Rapid cell growth (e.g., after vitamin B12 treatment for pernicious anemia)
Metabolic alkalosis In general, [K+]Plasma falls by 0.4 mmol/L per 0.1 U increase in ECF pH
Inhibition or blockade of K+ conductance Barium causes irreversible blockade of K+ conductance channels, preventing passive efflux of
channels and unopposed Na+/K+- ATPase K+ from cells
Chloroquine toxicity causes a dose-dependent reversible blockade of K+ channels
Familial hypokalemic periodic paralysis: K+ channels are normal but the muscle behaves as if
channels are blocked
Accelerated loss of potassium (can be classified according to acid-base status)
Chloride-responsive metabolic alkalosis Gastrointestinal loss (e.g., vomiting, gastric suctioning, colonic villous adenoma)
(chloride depletion causes ECF Renal loss (e.g., loop diuretics, Bartter’s syndrome, thiazide diuretics, Gitelman syndrome)
contraction with secondary
hyperaldosteronism, leading
to renal potassium wasting)
Chloride-resistant metabolic alkalosis Primary hyperaldosteronism: high aldosterone, low renin, normal cortisol levels, e.g., adrenal
(the primary disturbance is adenoma, bilateral adrenal hyperplasia
mineralocorticoid excess Glucocorticoid excess: normal aldosterone, normal renin and high cortisol levels, e.g., Cushing’s
resulting in sodium and water syndrome
retention, ECF expansion, Syndrome of apparent mineralocorticoid excess: low aldosterone level, low renin level, and
hypertension, metabolic normal cortisol, e.g., liquorice, Liddle syndrome
alkalosis, and the continuous Congenital adrenal hyperplasia: low aldosterone, low renin, and low cortisol
stimulation of potassium
secretion in the cortical
collecting duct)
Hyperchloremic metabolic acidosis Gastrointestinal loss of potassium, e.g., diarrhea, urinary diversion
(non anion gap) Renal tubular acidosis
Organic metabolic acidosis (high anion gap) E.g., lactic acidosis, ketoacidosis
Magnesium deficiency (normal acid-base E.g., alcoholism, malabsorption, drugs: diuretics, gentamicin, cisplatin, amphotericin
balance)
13. Clinical Hyperkalemia and Hypokalemia 75

Magnesium Depletion and upper extremities, with eventual impairment


of respiratory muscle function and ventilatory
Concurrent hypomagnesemia coexists in up to failure. Rhabdomyolysis can be seen with severe
40% of cases of hypokalemia because of drugs or hypokalemia.
disease processes that cause loss of both ions.
Whether hypokalemia is caused by magnesium
depletion or is an independent effect, it is difficult
fi Cardiovascular
to assess. Magnesium is essential in the stabiliza-
tion of cellular membranes, and hypomagnesemia Arrhythmias
is associated with increased losses of intracellular The arrhythmogenic potential of hypokalemia is
potassium, and increased fecal and urinary potas- significantly
fi increased in the presence of digoxin,
sium wasting. Regardless of the cause, hypokale- myocardial ischemia, congestive cardiac failure
mia becomes refractory when hypomagnesemia is (CCF), or left ventricular hypertrophy. Hypokale-
also present, especially with plasma magnesium mia predisposes to serious ventricular ectopy and
concentrations less than 0.6 mmol/L. In these cir- is associated with increased frequency of primary
cumstances, correction of hypokalemia requires ventricular fifibrillation and mortality, particularly
initial restoration of magnesium. in the setting of ischemic heart disease and acute
myocardial infarction.

Clinical Effects ECG Changes

Hypokalemia alters the function of excitable • Flattened T wave


membranes, an effect more notable in skeletal • U wave (delayed ventricular and Purkinje
muscle than cardiac muscle. Renal and metabolic repolarization)
effects are also seen. The likelihood of symptoms • Ventricular ectopics
seems to correlates with the rapidity of decrease • Note: When potassium is at ≤2.0 mmol/L, T and U
in plasma potassium. waves combine, creating a wave with greater
amplitude with risk of reentry circuits arising
during the prolonged relative refractory period,
Neuromuscular leading to the development of tachyarrhythmias

Mild Hypokalemia
Worsening of Cardiac Function
Mild hypokalemia (plasma potassium, 3.0–
Chronic hypokalemia exacerbates CCF by impair-
3.5 mmol/L) is often asymptomatic. Skeletal
ing the function and contractility of myocardial
muscle weakness is not usually seen with potas-
muscle. This reduces stroke volume and cardiac
sium greater than 3.0 mmol/L.
output. Cardiac necrosis has been described.
Moderate Hypokalemia
Symptoms of moderate hypokalemia (plasma Renal and Metabolic
potassium, 2.5–3.0 mmol/L) are nonspecific:fi lassi- Ammoniagenesis and Metabolic Alkalosis
tude, fatigue, constipation, weakness in the lower
extremities, and myalgia. A paralytic ileus can Chronic hypokalemia results in ICF acidification,

occur. which stimulates ammoniagenesis in the proximal
tubule. Increased ammonia trapping and net
acid excretion coupled to enhanced bicarbonate
Severe Hypokalemia
production perpetuates metabolic alkalosis.
Ascending paralysis can occur in severe hypoka- Increased ammoniagenesis may be clinically
lemia (plasma potassium, ≤2.0–2.5 mmol/L). The important in patients with severe liver disease,
lower extremities are typically involved fi
first (par- in whom hypokalemia may precipitate hepatic
ticularly the quadriceps), followed by the trunk encephalopathy.
76 H.-Y. Ong

Nephrogenic Diabetes Insipidus route of administration depending on the plasma


potassium as well as the presence and serious-
Chronic hypokalemia commonly leads to irrevers-
ness of the pathophysiological consequences of
ible tubulointerstitial changes resulting in neph-
hypokalemia. The underlying disorder should be
rogenic diabetes insipidus (DI).
treated simultaneously.
Continuous ECG monitoring of cardiac rhythm
Diagnostic Approach to Hypokalemia is indicated for plasma potassium ≤2.5 mmol/L or
intravenous infusion rates of at least 10 mmol/h.
1. History of associated conditions, including Maximum rate of intravenous potassium admin-
drug history. istration should not exceed 20 mmol/h. Large
2. Examination: assessment of extracellular enteral doses of potassium are difficult
fi to deliver
volume and acid-base status. because potassium salts are mucosal irritants,
3. Investigations: with both tablet and liquid forms reported to
a. Urinary potassium. cause esophageal and gastrointestinal ulceration.
i. Measured on a 24-hour urine collection,
urinary potassium should be less than 10
Suggested Reading
to 15 mmol/24 hours with hypokalemia
of extrarenall origin (indicating appro- Gennari FJ. Current Concepts: Hypokalemia. N Engl J
priate renal potassium conservation). Med. 1998; 339(7): 451–457.
ii. Random urinary potassium may be Glover P. Hypokalaemia. Crit Care Resusc. 1999; 1:
greater than 20 mmol/L in cases of renal 239–251.
Halperin M, Kamel KS. Potassium. Lancett 1998; 352:
potassium wasting, but this cannot be
135–140.
interpreted unambiguously because
Mahoney BA, Smith WAD, Lo DS, et al. Emergency Inter-
urinary potassium reflects
fl the K+ to H2O ventions for Hyperkalaemia. Cochrane Database Syst
ratio (and depends mainly on the amount Rev. 2006. Issue 1.
of water reabsorbed). Peterson LN, Levi M (Edited by Robert W. Schrier).
b. Urinary anion gap. Renal and Electrolyte disorders: Disorders of
4. Serum magnesium. Potassium Metabolism. 6th ed. Lippincott, Williams
5. Plasma aldosterone, renin, and cortisol levels. and Wilkins. 2003.
Rastegar A, Soleimani M. Hypokalaemia and hyperka-
laemia. Postgrad Med J. 2001; 77: 759–764.
Treatment
The immediate objective of potassium supple-
mentation is to prevent life-threatening cardiac
and muscular complications, with the rate and
14
Clinical Hyponatremia and Hypernatremia
Himangsu Gangopadhyay

Hyponatremia Transurethral Resection of the Prostate


(TURP) Syndrome
Hyponatremia reflects
fl an abnormal ratio of
• Hyponatremia
sodium to water and is defi
fined as a serum sodium
• Cardiovascular changes: hypertension/
less than 135 mmol/L. Mild to moderate hypona-
hypotension, bradycardia
tremia (serum sodium 128–134 mmol/L) is seen in
• Neurological changes: agitation, confusion,
20 to 30% of hospital inpatients. Severe hypona-
fitting, visual disturbances, fixed dilated
tremia (serum sodium less than 120 mmol/L) is
pupils
seen in only 2 to 5% of hyponatremic patients.
Irrigating fluid usually containing 1.5% glycine
is absorbed during the procedure. Excess fl fluid
Classification (Table 14.1) is absorbed and leads to an increase in total
body water and hyponatremia. There is an in-
1. Hyponatremia with decreased sodium and crease in the osmolar gap. With the use of
water (hypotonic dehydration). There is gener- glycine, other features may be seen: hypergly-
ally loss of sodium and water with inappropri- cinemia, hyperserinemia (a metabolite of
ate fluid replacement. glycine), hyperammonemia, metabolic acidosis,
2. Hyponatremia with normal body sodium and and hypocalcemia.
normal or increased body water, the patient is
usually euvolemic. Plasma osmolality may be Syndrome of Inappropriate Antidiuretic
normal, low, or high.
3. Hyponatremia with increased sodium and
Hormone Secretion
water. Syndrome of inappropriate antidiuretic hormone
(ADH) secretion (SIADH) is diagnosed on the fol-
lowing features:
Pseudohyponatremia • Hypotonic hyponatremia
Isosmolar hyponatremia (serum osmolality of • Urine osmolality greater than plasma osmolal-
275–290 mOsm/kg) is usually caused by severe ity (inappropriately concentrated urine
hyperlipidemia or hyperproteinemia and has osmolality)
been called “pseudohyponatremia.” Measurement • Urine sodium greater than 20 mmol/L
of plasma sodium by an ion-selective electrode is • Other causes of hyponatremia excluded (e.g.,
not affected by the volume of plasma “solids” and, cardiac, endocrine)
therefore, will give a true sodium level. • Normovolemia

77
78 H. Gangopadhyay

TABLE 14.1. Causes and classification of hyponatremia


Osmolality Volume status Other
Decreased sodium and water (hypotonic dehydration)
Extra renal sodium loss Excessive sweating Low Hypovolemic Urine Na < 20 mmol/L
Burns
Vomiting
Diarrhea
Renal sodium loss Osmotic diuretics Low Hypovolemic Urine Na > 20 mmol/L
Thiazide and loop diuretics
Mineralocorticoid deficiency
Salt-losing nephropathy
Diuretic phase of acute tubular
necrosis
Renal tubular acidosis
Normal body sodium and normal or increased body water
High plasma osmolality Hyperglycemia High Normal or hypovolemic
Mannitol
Glycerol
Glycine
Sorbitol
Normal plasma osmolality Hyperlipidemia Normal Normal
Hyperproteinemia
(pseudohyponatremia)
Low plasma osmolality Stress (physical, psychological) Low Normal Urine Na > 20 mmol/L
Antidiuretic drugs
Glucocorticoid deficiency
Myxedema
SIADH
Transurethral resection of prostate
Acute/chronic renal failure
Severe polydipsia Low Normal Urine Na < 20 mmol/L
Increased sodium and water
Edematous states Cardiac failure Low Edematous
Liver failure
Renal failure
Nephrotic syndrome

TABLE 14.2. Causes of SIADH


Ectopic ADH production Small cell bronchial carcinoma
Pancreatic adenocarcinoma
Leukemia
Lymphoma
Thymoma
Central nervous system disorders Trauma
Brain tumor
Central nervous system infection (meningitis, encephalitis,
abscess)
Subarachnoid hemorrhage
Acute intermittent porphyria
Guillain-Barré syndrome
Systemic lupus erythematous
Pulmonary diseases Pneumonia
Tuberculosis
Lung abscess
14. Clinical Hyponatremia and Hypernatremia 79

Treatment is with fluid restriction and treat- Asymptomatic Patients


ment of the underlying cause if possible. See Table
14.2 for a list of causes. It is important to fully assess the patient and treat
the underlying cause. The patient’s fluid fl status
should be assessed; hypovolemia should be treated
Clinical Presentation with normal saline, whereas hypervolemic states
(such as congestive cardiac failure) will require
Clinical presentation depends on the rapidity and salt and water restriction. With patients who are
severity of the development of the hyponatremia euvolemic and asymptomatic, fluid fl restriction
as well as the underlying condition. Mild may be adequate.
hyponatremia may be symptomless or present
with anorexia, nausea, lethargy, and apathy. Signs
and symptoms of severe hyponatremia include Symptomatic Patients
disorientation, agitation, seizures, depressed If the onset is acute and the patient is symptomatic
refl
flexes, focal neurological signs, and, eventually, (e.g., fitting, altered conscious level) then aggres-
Cheyne-Stokes respiration. sive correction of sodium is required. This can be
performed by the cautious infusion of 3% saline
at a rate of 1 mL/kg/h until the sodium rises to 120
Diagnosis to 125 mmol/L. The patient should be closely mon-
itored during this period, and the presence of
Diagnosis of the cause of hyponatremia involves: seizure or coma may require airway protection,
anticonvulsants, and other supportive care before
1. History: the serum sodium is corrected appropriately.
a. Medical history: cardiac disease, renal disease, Serum sodium should not be allowed to rise more
hepatic disease, endocrine disorders than 2 mmol/L/h and not more than 25 mmol in
b. Current and recent medication: diuretics, 48 hours. Frequent checking of serum sodium is,
antidepressants therefore, important. In patients with chronic
2. Physical examination: volume status, evidence hyponatremia, a more gradual rise of serum
of cardiac, renal, liver failure sodium is appropriate.
3. Investigations:
a. Serum osmolality
b. Urine sodium and osmolality
c. Blood glucose
Hypernatremia
d. Serum lipids
Hypernatremia is defined
fi as serum sodium greater
e. Thyroid function
than 145 mmol/L. It is always associated with a
f. Plasma proteins
hyperosmolar state.

Treatment
Classification (Table 14.3)
Much has been mentioned regarding treatment of
hyponatremia and the potential complication of 1. Decreased total body sodium (extracellular
central pontine myelinolysis (CPM). This is a water and sodium loss with excess water
potentially fatal condition that can occur when loss)
the serum sodium is corrected too quickly. CPM 2. Normal total body sodium (extracellular water
can lead to mutism, dysphasia, spastic quadriple- deficiency
fi associated with minimal sodium
gia, pseudobulbar palsy, delirium coma, and even loss)
death. Risk factors for its development are alco- 3. Increased total body sodium (extracellular
holism, burns, use of thiazide diuretics, a mal- water and sodium gain with relatively excess
nourished state, and hypokalemia. sodium gain)
80 H. Gangopadhyay

TABLE 14.3. Classification and causes of hypernatremia Evaluation of Hypernatremia


Decreased total body sodium (water greater than sodium loss)
Extra renal loss Vomiting Diagnosis of the cause of hypernatremia
Diarrhea involves:
Excessive sweating
Dialysis 1. History
Renal loss Osmotic diuretics (e.g., glucose, urea, a. Fluid loss/excessive water loss
mannitol)
b. Poor fluid intake
Normal total body sodium (water loss) c. Increased salt intake
Extra renal Unconscious state
d. Intravenous sodium bicarbonate or hyper-
Thirst center dysfunction
Mechanical obstruction tonic saline therapy
Inappropriate intravenous therapy e. Clinical scenario that can lead to neuro-
No access to water genic/nephrogenic diabetes insipidus
Renal loss Cranial diabetes insipidus 2. Physical examination: assessment of volume
Nephrogenic diabetes insipidus
status is most important
Increased total body sodium (sodium greater than water gain) 3. Investigations:
High sodium intake Sea water ingestion a. Serum osmolality
Accidental/intentional salt ingestion
Hypertonic saline
b. Urine sodium and osmolality
Sodium bicarbonate infusion
Low sodium output Mineralocorticoid excess
Interpretation of Urinary Osmolality
A serum osmolality greater than 290 mOsm/kg
should be accompanied by highly concentrated
urine if the concentrating mechanism of the
Mechanism tubules is intact and ADH release is adequate. A
urinary osmolality of less than 200 mOsm/kg
The defence mechanisms against hypernatremia usually suggests some form of diabetes insipidus.
are ADH release and thirst. Any clinical condition
affecting these mechanisms will lead to hyperna-
tremia if not addressed properly. Treatment
Treating underlying cause is very important. Fluid
Clinical Presentation therapy with pure water orally or nasogastrically
could be suffi
ficient in some situations, but intrave-
Clinical signs and symptoms are not seen until nous therapy with 5% dextrose or pure water
serum sodium reaches more than 155 mmol/L. through a central vein is often necessary.
Symptoms include fever, restlessness, irritability, A rapid decrease of serum sodium could be
drowsiness, lethargy, confusion, and coma. Con- equally detrimental, and a decrease of serum
vulsions are rarely seen. sodium by 2 mmol/L/h is acceptable.
15
Clinical Metabolic Acidosis and Alkalosis
Sara Blakeley

Acid-base disturbances are common on the inten- if one or more of these three variables changes,
sive care unit (ICU) and should always prompt a rather than bicarbonate itself being the main deter-
search for the underlying cause (Tables 15.1–15.3). minant of change, as in the traditional approach.
Recently another, although not all that new (1), “Strong ions” are ions that exist completely dis-
way of understanding the mechanism behind sociated (charged) in water. There are more strong
acid-base disorders has gained popularity. This cations than anions and this difference is called
chapter starts with just a brief discussion of this the SID. SID has an electrochemical effect on water
approach, because there are several excellent over- dissociation; as SID becomes more negative, H+
views in the literature (2–6). increases and pH falls.
“Weak ions” exist as dissociated (A−) or associ-
ated forms (HA), and together they make up ATOT.
Traditional Versus Physiochemical Nonvolatile weak acids, mainly albumin but also
Approach to Acid-Base Disturbances inorganic phosphate and bicarbonate, dissociate
in body fluids forming weak anions.
Most “traditional” views of acid-base balance rely To maintain electrical neutrality, the SID must
on the Henderson-Hesselbach equation, but there be balanced by an equal and opposite charge,
has always been debate regarding quantification
fi the effective SID (SIDe). The SIDe is made up of
of the metabolic component. It can be described predominately weak acids (A−) and CO2 (Equation
using plasma bicarbonate and anion gap (AG) or 15.1). If the SID is calculatedd from the measured
by using standard base excess (SBE). However, plasma strong ion concentrations (Equation 15.2),
SBE is affected by changes in sodium, chloride, it is termed the apparent SID (SIDa). However, this
and albumin, and a significant
fi number of abnor- will not be precise if there are unmeasured anions
mal AGs will be missed unless the effect of albumin or cations present. If the SIDe and the SIDa do not
is considered (7–9). The third way of quantifying match (strong ion gap [SIG]) then unmeasured
the metabolic component is the strong ion differ- ions mustt be present. SIG should be zero, although,
ence (SID) (1), taking into consideration nonbi- in practice, there is a degree of normal variability.
carbonate buffers, such as inorganic phosphate If SIG is positive, there must be more unmeasured
and serum albumin, both of which may be abnor- strong anions (e.g., ketones, sulfates), and if it is
mal in critically ill patients. negative, more unmeasured cations.
Stewart’s (physiochemical) approach relies on
Equation 15.1:
the principles of physical chemistry; electroneu-
SIDe = A− + HCO3−
trality, conservation of mass, and dissociation of
electrolytes (1). It considers three independent Equation 15.2:
variables for pH: the SID, pCO2 and total weak acid SIDa = ([Na+] + [K+] + [Ca2+] + [Mg2+]) − ([Cl−] +
concentration (ATOT). H+ or HCO3− can only change [lactate])

81
82 S. Blakeley

TABLE 15.1. Classification of metabolic acidosis according to AG


High AG (unmeasured anions)
Lactate
Ketones Diabetic, alcoholic, or starvation ketoacidosis
Renal failure
Ingested toxins E.g., salicylate, paracetamol, formaldehyde, ethylene glycol, methanol
Non-AG (hyperchloremic)
Gastrointestinal bicarbonate loss E.g., diarrhea, ureterosigmoidostomy, jejunal/ileal loop, small bowel fistulae, pancreatic
fistulae, biliary fistulae
Renal bicarbonate loss Renal tubular acidosis
Exogenous acid load
Loss of potential bicarbonate E.g., ketosis with ketone excretion
Cation exchange resins
Low AG
Hypoalbuminemia
Increased concentration of unmeasured cations E.g., magnesium, calcium, lithium
Accumulation of positively charged compounds E.g., cationic antibiotics, IgG myeloma
normally absent from serum
Interference with measurement E.g., hyperlipidemia
+ + − −
AG = ([Na ] + [K ]) + ([Cl ] + [HCO ]) (Normal, 10–18 mmol/L)
3
AGadjusted = AGobserved + 0.25 × ([normal albumin in g/L] − [observed albumin in g/L])

TABLE 15.2. Pathological classification of causes of metabolic alkalosis


Exogenous or endogenous bicarbonate load
Acute alkali administration
Milk alkali syndrome
Recovery from lactic acidosis or ketoacidosis Caused by excess regeneration of HCO3
Massive blood transfusion Caused by metabolism of citrate
Chloride depletion
Vomiting, high gastric drainage
Gastrocystoplasty
Diuretics
Posthypercapnia
Diarrhea secondary to villous adenoma Villous adenomas are usually associated with hyperchloremic metabolic acidosis, but some can
secrete chloride
Cystic fibrosis
Potassium deficiency
Primary hyperaldosteronism Caused by adenoma, carcinoma; idiopathic or hyperplasia
Secondary hyperaldosteronism E.g., Cushing syndrome/disease, exogenous corticosteroids, accelerated hypertension, renal
artery stenosis
Drugs E.g., carbenoxolone, liquorice
Primary hyperaldosteronism Caused by adenoma, carcinoma; idiopathic or hyperplasia
Secondary hyperaldosteronism E.g., Cushing syndrome/disease, exogenous corticosteroids, accelerated hypertension, renal
artery stenosis
Bartter’s syndrome
Gitelman syndrome
Magnesium or potassium deficiency
High-dose intravenous penicillins Caused by distal delivery of nonreabsorbable anions with an absorbable cation such as sodium
Other
Severe hypoalbuminemia E.g., nephrotic syndrome
15. Clinical Metabolic Acidosis and Alkalosis 83

TABLE 15.3. Clinical effects of metabolic acidosis and metabolic alkalosis


Metabolic acidosis Metabolic alkalosis
Cardiac • Reduced myocardial contractility and cardiac failure • Decreased myocardial perfusion caused by arteriolar
(but unclear association in clinical practice) constriction
• Resistance to catecholamines • Reduced angina threshold
• Increased risk of arrhythmias • Increased risk of arrhythmias
Respiratory • Stimulation of the carotid body and aortic • Depressed ventilation (causing compensatory respiratory
chemoreceptors causing hyperventilation acidosis), which could lead to failure to wean a patient
(Kussmaul respiration) from mechanical ventilation
Neurological • Impaired consciousness (mechanism not fullyy • Decreased cerebral blood flow caused by arteriolar
understood and not clearly correlating with the constriction
degree of acidosis) • Headache, confusion, mental obtundation
• Intracerebral vasodilation (may be clinicallyy • Neuromuscular excitability: seizures and tetany (related to
significant in the setting of already raisedd reduction in ionized calcium level)l
intracranial pressure)
Metabolic and immune • Hyperkalemia (extracellular potassium shifts) • Hypokalemia
• Glucose intolerance (inhibition of glycolysis andd • Hypomagnesemia
hepatic gluconeogenesis)
• Immune activation • Hypophosphatemia
• Stimulation of aerobic glycolysis leading production of
lactic acid and ketoacids
• Decreased plasma ionized calcium concentration

Note: the effects of severe alkalemia (pH > 7.60) appear more pronounced with respiratory rather than metabolic alkalosis.

Equation 15.3: lactate can be produced under aerobic conditions,


SIDa − SIDe = SIG (equals zero unless especially in the setting of infl flammatory pro-
unmeasured anions or cations cesses. In sepsis, infl flammatory mediators can
present) accelerate aerobic glycolysis, leading to increased
glucose turnover and increased lactate production
By using this approach, a change in the balance
(12, 13). Specifific organs are also capable of pro-
between strong anions and cations accounts for
ducing excess lactate under times of stress, for
acid-base disturbances seen on the ICU. For
example, the lungs and gut (12, 13).
example, excess chloride from a high-volume
Elevated lactate levels may also indicate
saline infusion reduces the SID, causing an acido-
decreased clearance (12). Lactate is mostly metab-
sis. A loss of chloride from a large nasogastric
olized by the liver (50%), and decreased clearance
output increases the SID, causing an alkalosis.
can occur with impaired liver function or
The modern debate now centers on which is the
decreased liver blood flow.
best and most practical method of assessing acid-
Unless lactate levels are specifi
fically measured,
base disorders in critically ill patients, and, in par-
an elevated level may be “missed” by the AG or
ticular, detecting the presence of unmeasured anions:
SBE because of factors such as hypoalbuminemia.
the SIG. To date, neither the SIG, corrected AG, or
In addition, hyperlactatemia is not always accom-
corrected SBE has been found to be overwhelmingly
panied by an acidosis.
superior to the other, but all have been found to be
Elevated lactate levels, particularly those that
predictors of increased mortality (10, 11).
fail to clear during the fifirst 24 hours, have been
repeatedly shown to correlate with a poor outcome
Lactic Acidosis (Table 15.4) (11, 14, 15).

When more lactate is produced than is metabo-


Use of Sodium Bicarbonate
lized, hyperlactatemia occurs. Elevated levels are
generally taken to indicate anaerobic metabolism Controversy exists regarding the use of alkali
caused by generalized or regional tissue hypoxia therapy (e.g., sodium bicarbonate) to treat meta-
(12). However, it is important to remember that bolic acidosis (16). No clinical studies have shown
84 S. Blakeley

TABLE 15.4. Classification of lactic acidosis


Type A (hypoxic)
Circulatory insufficiency (e.g., shock, cardiac arrest)
Regional hypoperfusion
Carbon monoxide and cyanide poisoning (caused by mitochondrial enzyme inhibition)
Severe hypoxia
Severe exercise/prolonged seizures (caused by increased muscle activity)
Severe anemia
Type B (nonhypoxic)
Sepsis in general (caused by accelerated aerobic glycolysis and mitochondrial dysfunction) and with specific infections (e.g., cholera, falciparum
malaria, AIDS)
Thiamine deficiency (thiamine is a cofactor of pyruvate dehydrogenase)
Fulminant hepatic failure, severe liver disease
Alcoholism (ethanol oxidation increases the conversion of pyruvate to lactate and inhibits of pathways of pyruvate metabolism)
Severe renal failure
Malignancy, leukemia, and lymphoma
Short bowel syndrome (D lactic acidosis)
Diabetic ketoacidosis (ketones may inhibit hepatic lactate uptake)
Type B: drug induced
Phenformin/metformin
Ethanol, methanol, ethylene glycol
Salicylate poisoning
Paracetamol poisoning
Intravenous fructose, sorbitol
Cyanide
β-agonists (e.g., salbutamol, adrenaline, caused by increased glyconeogenesis, glycogenolysis, lipolysis, and cyclic AMP activity)
Type B: inborn errors of metabolism
Glucose 6 phosphatase deficiency
Fructose 1,6 diphosphatase deficiency
Deficiency of enzymes of oxidative phosphorylation

a benefifit on outcome in patients with lactic acido- cacy of renal replacement therapy (RRT) tech-
sis, but it is diffi
ficult to separate the effects of the niques, therefore, the acidosis can be cleared
acidosis from the effects of the underlying patho- within 48 hours (16). Despite the fact that lactate
logical condition. has a low molecular weight and it is easily removed
Sodium bicarbonate is not without risks, by continuous therapies, RRT has been shown to
including: contribute to only less than 3% of total lactate
clearance (17). It may be that RRT plays a role in
• Acute fluid overload. improving the overall clinical situation (e.g.,
• Alkaline “overshoot” caused by subsequent hemodynamic and fluid fl status), therefore, pro-
metabolism of organic salts increasing the bicar- moting the clearance of lactate, rather than actu-
bonate concentration. ally removing it.
• Paradoxical intracellular acidosis. CO2 released
diffuses more quickly than bicarbonate ions into
cells, therefore, initially lowering the intracellu-
lar pH. There is debate regarding whether this is Metabolic Alkalosis
clinically significant.

Metabolic alkalosis is common, but carries an
associated mortality that increases with increas-
Use of Renal Replacement Therapy ing pH, especially pH higher than 7.55 (18). The
In acute renal failure, the daily production of cause, rather than the alkalosis itself, may be the
mineral acids is adequately matched by the effi-
fi reason for the increased mortality.
15. Clinical Metabolic Acidosis and Alkalosis 85

Metabolic alkalosis can be classified


fi clinically 3. Consider specificfi treatments, e.g., surgery
(Table 15.2) or according to the underlying for an adrenal adenoma or spironolactone for
pathophysiology: initiating process or perpetuat- hyperaldosteronism.
ing processes (18). 4. Other:
a. Hydrochloric acid: rarely used.
b. Acetazolamide: increases bicarbonate loss,
Initiating Processes but only if GFR is adequate.
c. RRT: in patients with severe cardiac or renal
1. Loss of hydrogen ions through the kidney
disease, RRT may assist volume and electro-
(e.g., loop diuretics, primary hyperaldosteronism)
lyte correction. Standard replacement flfluids
or gastrointestinal tract (e.g., vomiting, nasogas-
will need modifification as they contain bicar-
tric suctioning).
bonate or its precursor lactate.
2. Accumulation of alkali from exogenous (e.g.,
sodium bicarbonate in the setting of renal insuf-
ficiency) or endogenous sources (e.g., metabolism
fi References
of ketones after diabetic ketoacidosis).
1. Stewart PA. Modern quantitative acid-base chemis-
The kidneys have a large capacity to excrete try. Can J Pharmacol. 1983; 61: 1444–1461.
excess bicarbonate and are mostly able to correct 2. Story DA. Bench-to-bedside review: A brief
the alkalosis; however, in certain situations, the history of clinical acid-base. Crit Care. 2004; 8:
kidney will retain rather than excrete alkali, there- 253–258.
fore, perpetuatingg the alkalosis. The persistence of 3. Kellum JA. Determinants of blood pH in health and
disease. Crit Care. 2000; 4: 6–14.
a metabolic alkalosis suggests there is an ongoing,
4. Kaplan LJ, Frangos S. Clinical review: Acid-base
untreated process. abnormalities in the intensive care unit. Crit Care.
2005; 9: 198–203.
Perpetuating Factors 5. Morgan TJ. Clinical review: The meaning of acid-
base abnormalities in the intensive care unit—
1. Hypovolemia. Decreased renal perfusion stimu- effects of fl
fluid administration. Crit Care. 2005; 9:
lates the renin-angiotensin-aldosterone system 204–211.
(RAAS), therefore, increasing sodium absorp- 6. Morgan TJ. What exactly is the strong ion gap, and
tion and hydrogen ion secretion. does anybody care? Crit Care Resusc. 2004; 6: 155–
2. Chloride depletion. Stimulation of the RAAS 166.
leads to increased bicarbonate reabsorption in 7. Figge J, Jabor A, Kazda A, Fencl V. Anion gap and
hypoproteinaemia. Crit Care Med. 1998; 26: 1807–
the absence of adequate chloride.
1810.
3. Severe hypokalemia. Caused by an intracellu- 8. Fencl V, Jabor A, Kazda A, Figge J. Diagnosis of
lar shift of hydrogen ions in exchange for metabolic acid-base disturbances in critically ill
potassium. patients. Am J Resp Crit Care Med. 2000; 162:
4. Reduced glomerular fi filtration rate (GFR). 2246–2251.
Reduction in GFR causes a decrease in filtered
fi 9. Story DA, Poustie S, Bellomo R. Estimating unmea-
bicarbonate. sured anions in critically ill patients: anion gap,
base deficit
fi and strong ion gap. Anaesthesia. 2002;
57: 1102–1133.
Treatment 10. Kaplan LJ, Kellum JA. Initial pH, base deficit,
fi lactate,
1. Correct the primary cause if possible, e.g., anion gap, strong ion difference, and strong ion gap
predict outcome from major vascular injury. Crit
stop or reduce diuretics.
Care Med. 2004; 32: 1120–1124.
2. Address perpetuating factors. Most forms of 11. Smith I, Kumar P, Molloy S, et al. Base excess and
metabolic alkalosis are chloride responsive, and lactate as prognostic indicators for patients ad-
isotonic sodium chloride will correct both chlo- mitted to intensive care. Intensive Care Med. 2001;
ride and volume depletion. As the chloride deficitfi 27: 74–83.
is corrected, there will be an alkaline diuresis and 12. De Backer D. Lactic acidosis. Intensive Care Med.
the plasma bicarbonate will start to normalize. 2003; 29: 699–702.
86 S. Blakeley

13. Bellomo R, Ronco C. The pathogenesis of lactic aci- 16. Levraut J, Grimaud D. Treatment of metabolic
dosis in sepsis. Curr Opin Crit Care. 1999; 5: 452–457. acidosis. Curr Opin Crit Care. 2003; 9: 260–
14. Stacpoole PW, Wright EC, Baumgartner TG, et al. for 265.
the DCA-Lactic acidosis Study Group: Natural 17. Levraut J, Ciebiera JP, Jambou P, et al. Effect of con-
history and course of acquired lactic acidosis in tinuous venovenous hemofiltration
fi with dialysis on
adults. Am J Med. 1994; 97: 47–54. lactate clearance in critically ill patients. Crit Care
15. Husain FA, Martin MJ, Mullenix PS, et al. Serum Med. 1997; 25(1): 58–62.
lactate and base defi ficit as predictors of mortality 18. Galla JH. Metabolic alkalosis. J Am Soc Nephrol.
and morbidity. Am J Surg. 2003; 185(5): 485–491. 2000; 11: 369–375.
Index

A Aneurysms, abdominal, 22, 35 Calcium channel antagonists, 17


N-Acetylcysteine, 17, 28, 36
N Angiotensin, 2 Calculi, ureteric, imaging studies
Acid-base disturbances. See also Angiotensin-converting enzyme of, 9–10
Acidosis; Alkalosis inhibitors (ACEIs), 14, 16, Cardiac arrest, hyperkalemia-
traditional versus 33 related, 71, 73
physiochemical approach Angiotensin receptor blockers, Cardiac output, 29
to, 81–84 nephrotoxicity of, 16 Cardiac surgery, as acute renal
Acidosis, 22, 39 Anion gap, 81, 82, 83 failure risk factor, 34–35
lactic, 83–84 Anticoagulation, in renal Casts, 6, 23, 26, 39
metabolic, 60, 82, 83, 84 replacement therapy, 61–63 Cation exchange resins, 73
Acute kidney injury (AKI), 19–25 Antidiuretic hormone (ADH), 4, Central pontine myelinolysis, 79
causes of, 20–22 80 Chemical testing, of urine, 4–5
investigation of, 23, 24 Anuria, 4, 19, 27 Chloride depletion, 82, 83
complications of, 22–23 Aprotinin, as acute renal failure Coagulopathy, therapeutic plasma
defifinition of, 19 risk factor, 34 exchange-related, 50
incidence and outcome of, 19–20 Aspartate transaminase, 38 Cockroft and Gault formula, for
Acute tubular necrosis (ATN), 3, 20, Azathioprine, 68 creatinine clearance, 3
22 Azotemia, 42 Compartment syndromes, 20, 34,
casts in, 6, 23 38–39, 41
imaging of, 7–9 B Complement, 44, 46
as oliguria cause, 27 Bacteriuria, 5 Compression injuries, as
as renal failure cause, 7, 21, 26, Bicarbonate, 40, 51, 60, 73, 83–84, rhabdomyolysis cause, 38
27–28 84, 85 Computed tomography (CT), renal,
urea-to-creatinine ratio in, 2–3 Biopsy, renal 7, 9–11
Adenosine agonists, 28 for acute renal failure diagnosis, Contrast agents, nephrotoxicity of,
Alcohol, as rhabdomyolysis cause, 27 10, 11, 13, 16–17, 27–28, 36
39 “poor man’s,” 3 Corticosteroids, 68
Alkalosis, metabolic, 74, 75, 82, for vasculitis diagnosis, 43 Creatine kinase, 38
84–85 Blood flow, renal Creatinine, 2–3
Allopurinol, 15 effect of prostaglandins on, 14 as acute kidney injury marker,
Aminoglycosides, nephrotoxicity regulation of, 33 19
of, 16, 33 in rhabdomyolysis, 38, 39–40 renal failure-related increase in,
Ammoniagenesis, 75 24
Anaphylaxis, dialysis-related, 67 C Creatinine clearance, 3
Anemia, acute kidney injury- Calcifi
fication, heterotopic, 39 Crush injuries, as rhabdomyolysis
related, 22, 24 Calcium. See also Hypercalcemia; cause, 38
Anesthesia, effect on renal Hypocalcemia Cyclooxygenase inhibitors, 15
function, 33 as hyperkalemia treatment, 73 Cyclosporin, 68

87
88 Index

D F Hypocalcemia, 39, 50
Daclizumab, 69 Familial hypokalemic periodic Hypokalemia, 50, 74–76, 82, 85,
Dextrose, 17, 73 paralysis, 74 8575, 73076
Diabetes insipidus, nephrogenic, 76 Fasciotomy, 38 Hypomagnesemia, 75
Diabetes mellitus, 6, 33, 34 Fluid composition, of the body, 1, Hyponatremia, 22, 77–79, 79
Dialysis 2 Hypoperfusion, renal, 9, 20.6
optimal dose in, 65–67 Fluid resuscitation, in acute renal Hypotension
peritoneal, 64–67 failure patients, 28–29 acute renal failure-related, 26, 29
principles of, 64 anesthesia-related, 33
as renal vasculitis treatment, 44 G dialysis-related, 67
for urinary myoglobin Glomerular filtration,
fi cessation of, as postoperative renal failure
reduction, 39–40 33 cause, 33
Dialysis equilibrium syndrome, 67 Glomerular filtration
fi rate (GFR), 2 prerenal failure-related, 20
Diethylene triamine pentaacetic in acute kidney injury, 19 relative, 36
acid (DTPA) studies, 7, 10 assessment of, 3 in surgical patients, 33
Digoxin, toxicity of, 73 in metabolic alkalosis, 85 therapeutic plasma exchange-
1,25-Dihydroxyvitamin D3, 1 Glomerulonephritis, 6, 9, 26 related, 50
Disseminated intravascular Glucose control, tight, 29, 36 Hypovolemia, 21, 49, 85
coagulation, 46–47 Glycosuria, renal, 5
Diuretics Goodpasture’s syndrome, 9 I
as acute renal failure Imaging, of acute renal failure,
prophylaxis, 34, 36 H 7–13. See also Computed
as acute renal failure treatment, Hematuria, 4, 26, 42 tomography (CT); Magnetic
36 Hemodiafiltration,
fi 64 resonance angiography
loop, 15, 17, 27–28 continuous venovenous, 59 (MRA); Magnetic resonance
nephrotoxicity of, 16 Hemodialysis, 64 imaging (MRI);
Dopamine, as renal failure complications of, 67 Scintigraphic imaging
prophylaxis, 36 continuous, 57–63 diagnostic algorithm for, 12
Dopamine agonists, 28 as hyperkalemia treatment, 74 Immunosuppression, renal failure-
Drugs. See also names of specificfi intermittent, 52, 54, 60 related, 22
drugs Hemofi filters, 50 Immunosuppressive therapy, in
as hypokalemia cause, 74 Hemofi filtration, 52, 64 renal transplant recipients,
impaired renal clearance of, 23–24 continuous venovenous, 52, 57, 68–69
nephrotoxicity of, 14–18, 23, 59 Infection
33–34, 38, 39 high-volume, 53 in renal transplant recipients, 69
as rhabdomyolysis cause, 38, 39 prophylactic, 29 as rhabdomyolysis cause, 38
for urinary myoglobin Inotrope therapy, for acute renal
E reduction, 39–40 failure, 29
Edema Hemofi filtration machines, 58, Insulin therapy, 36, 73
pulmonary, 43 59–60 Intensive care unit (ICU) patients
renal, 9 Hemolytic uremic syndrome, 46, acute kidney injury in, 19
Electrocardiography 47 acute renal failure in, 26
in hyperkalemia, 71–72 Hemorrhage, gastrointestinal, 23 intrinsic renal failure in, 20, 21
in hypokalemia, 75 Henoch-Schönlein purpura, 43 obstructive renal failure in, 22
Electrolyte disturbances. See also Heparin, 61–62 Inulin clearance, 3
specific
fi electrolyte Homeostasis, role of the kidney in, Ischemia, renal, 33
disturbances 1
acute kidney injury-related, 22 Hypercalcemia, 24 K
Encephalopathy, uremic, 22 Hyperglycemia, 22 Kidney
Enteral support, in critically ill Hyperkalemia, 16, 22, 71–73 hydronephrotic, 9
patients, 29 Hypernatremia, 79–80 normal, ultrasound imaging of,
Eosinophilia, 24 Hyperphosphatemia, 22, 24, 39 7, 8
Erythropoietin, 1, 22 Hypertension, 13, 42 normal functions of, 1
Excretion, urinary, 1 malignant, 47 polycystic, 7
Index 89

L Pregnancy, in systemic lupus Renal function


Lactate, 60, 82, 83 erythromatosus patients, 46 abrupt and sustained decrease
Lactate dehydrogenase, 38 Prerenal failure, 7, 21 in. See Acute kidney injury
Left ventricular end diastolic as acute kidney injury cause, 20 (AKI)
pressure (LVEDP), 29 defifinition of, 20 assessment of, 1–6, 26
differentiated from intrinsic Renal index (RI), 9
M renal failure, 23, 24 Renal injury. See also Acute kidney
Magnetic resonance angiography Prostacyclin, 63 injury (AKI)
(MRA), renal, 10–11 Prostaglandins, renal, 14–15 drug-induced, 14–18
Magnetic resonance imaging Proteinuria, 4–5 Renal replacement therapy, 28,
(MRI), renal, 7 Pseudohyperkalemia, 72–73 51–56. See also Dialysis;
Mannitol, 28, 34 Pseudohyponatremia, 77 Hemodialysis; Renal
Mean arterial pressure (MAP), 29, Purpura, thrombotic transplantation
33 thrombocytopenic, 46, 47 for acute kidney injury, 20
Mercaptoacetyltriglycerine (MAG3) anticoagulation in, 62
studies, 7, 8, 9, 10, 11 R basics of, 51–52
Microangiopathy, thrombotic, Red blood cells, in urine, 6 continuous, 51, 52, 53, 54, 57–59,
46–47 Renal artery, dissection or 61
Mineral disturbances, acute kidney occlusion of, 10–11, 13, 16 for hyperkalemia, 74
injury-related, 22 Renal disease, end-stage, renal effect on lactic acidosis, 84
Mycophenolate mofetil, 69 replacement therapy for, for end-stage renal disease, 64–70
Myoglobin, in urine, 39–40 64–70 hemofi filters in, 60
Renal dysfunction. See also Acute hybrid, 52
N kidney injury (AKI) indications for, 53–54
Natriuretic peptides, 28, 36 RIFLE classifi fication system for, intermittent, 51, 53, 54
Nephritic syndrome, 42–43 19, 20 replacement and dialysis fluid

Nephritis Renal failure in, 59–60
interstitial, 6, 9, 15, 33–34 acute selection of dosage and mode in,
systemic lupus erythromatosus- acute kidney injury-related, 19 54–55
related, 45 diagnosis of, 26, 35–36 technical aspects of, 57–63
Nephrotic syndrome, 5, 11, 16 differentiated from chronic vascular access in, 52, 60–61
Neutrophilia, 24 renal failure, 7 for volume overload, 22
Nonsteroidal anti-infl flammatory drug-related, 14–18 Renal transplantation, 67–69
drugs, nephrotoxicity of, imaging of, 7–13 Renal vein, thrombosis of, 11, 13
14–16, 33 laboratory investigations of, Renin, 1, 2
Nutrition/nutritional support, 29, 26–27 Respiratory failure, 43
67 medical management of, Resuscitation, in surgical patients,
26–32 36
O multisystem causes of, 42–48 Rhabdomyolysis, 28, 38–41
Oliguria, 4, 27, 34 pathophysiology of, 33 diagnosis of, 38–39
Oxygen consumption, renal, 33 prevention of, 27–30, 36–37 drug-related, 14
rhabdomyolysis-related, 38 treatment of, 40
P risk factors for, 26, 27, 33–34 RIFLE system, for renal
Peritoneal dialysis, 64–67 supportive strategies for, 28–29 dysfunction classification,

Plasma exchange, therapeutic, in surgical patients, 33–37 19, 20
49–50, 59 treatment of, 35–36
Plasma volume, calculation of, 49 chronic S
Polyclonal antibodies, 69 differentiated from acute renal Salbutamol, 73
Polyuria, 4 failure, 7 Scintigraphic imaging
Postrenal failure, 7 imaging of, 8 diethylene triamine pentaacetic
Potassium. See also Hyperkalemia; intrinsic, 20–21 acid (DTPA) studies, 7, 10
Hypokalemia differentiated from prerenal mercaptoacetyltriglycerine
renal excretion of, 71, 73 failure, 23, 24 (MAG3) studies, 7, 8, 9, 10, 11
Potassium supplementation, 76 postrenal or obstructive, 22 of ureteric obstruction, 10
90 Index

Sepsis of renal vein thrombosis, 11 Urological surgery, as acute renal


dialysis-related, 67 of ureteric obstruction, 9 failure risk factor, 35
treatment of, 53 Urea
Sodium bicarbonate, 40, 73, 84 accumulation of, 22, 24 V
Statins, as rhabdomyolysis cause, measurement of, 2 Vascular surgery, as acute renal
39 Urea-to-creatinien ratio, 2–3 failure risk factor, 34
Strong ion difference (SID), 81, 83 Uremia, 22–23 Vasculitis
Surgical patients Uremic patients, cardiovascular as acute renal failure cause, 26,
acute kidney injury in, 20 risk factors in, 65 42–45
acute renal failure in, 33–37 Ureteric obstruction, imaging clinical presentation of, 42–43
Syndrome of inappropriate studies of, 9–10 etiology of, 42
antidiuretic hormone Urinalysis, 3–6 treatment of, 43–44
secretion (SIADH), 77, 78, for acute kidney injury Wegener’s renal, 44
79 evaluation, 23, 24 Venography, of renal vein
Systemic lupus erythromatosus, for acute renal failure evaluation, thrombosis, 11
45–46 26–27 Volume depletion
for glomerulonephritis/acute as acute renal failure cause, 26
T interstitial nephritis assessment and correction of, 29
Tacrolimus, 68 differentiation, 9 as postoperative renal failure
Tamm-Horsfall protein, 40 in surgical patients, 35 cause, 33
Thrombocytopenia, 24 for systemic lupus prerenal failure-related, 21
Toxins erythromatosus evaluation, Volume expansion, intravascular,
as rhabdomyolysis cause, 38 45 40
uremic, retention of, 22 Urine Volume overload, acute kidney
Transurethral resection of the alkalinization of, 14, 40, 41 injury-related, 22
prostate (TURP) syndrome, appearance of, 3
77 chemical testing of, 4–5 W
Tubulointerstitial disease, 26 microscopy of, 5–6 Wegener’s granulomatosis, 9
Tumor lysis syndrome, 14 in rhabdomyolysis, 39, 40 Wegener’s renal vasculitis, 44
osmolality of, 23, 24, 80 White blood cells, in urine, 5–6
U specifific gravity of, 3, 24
Ultrafi
filtration, slow continuous, 53, volume of, 3–4, 27 X
59 Urine output, in acute kidney X-rays
Ultrasound, renal, 7, 23, 27 injury, 19–20 chest, 23, 44
of normal kidneys, 7, 8 Urolithiasis, 27 renal, 7

You might also like