Oxford Handbook of Endocrinology and Diabetes 3rd Ed
Oxford Handbook of Endocrinology and Diabetes 3rd Ed
Oxford Handbook of Endocrinology and Diabetes 3rd Ed
Oxford Handbook of
Endocrinology and
Diabetes
Published and forthcoming Oxford Handbooks
Oxford Handbook for the Foundation Programme 3e
Oxford Handbook of Acute Medicine 3e
Oxford Handbook of Anaesthesia 3e
Oxford Handbook of Applied Dental Sciences
Oxford Handbook of Cardiology 2e
Oxford Handbook of Clinical and Laboratory Investigation 3e
Oxford Handbook of Clinical Dentistry 5e
Oxford Handbook of Clinical Diagnosis 2e
Oxford Handbook of Clinical Examination and Practical Skills
Oxford Handbook of Clinical Haematology 3e
Oxford Handbook of Clinical Immunology and Allergy 3e
Oxford Handbook of Clinical Medicine – Mini Edition 8e
Oxford Handbook of Clinical Medicine 9e
Oxford Handbook of Clinical Pathology
Oxford Handbook of Clinical Pharmacy 2e
Oxford Handbook of Clinical Rehabilitation 2e
Oxford Handbook of Clinical Specialties 9e
Oxford Handbook of Clinical Surgery 4e
Oxford Handbook of Complementary Medicine
Oxford Handbook of Critical Care 3e
Oxford Handbook of Dental Patient Care
Oxford Handbook of Dialysis 3e
Oxford Handbook of Emergency Medicine 4e
Oxford Handbook of Endocrinology and Diabetes 3e
Oxford Handbook of ENT and Head and Neck Surgery 2e
Oxford Handbook of Epidemiology for Clinicians
Oxford Handbook of Expedition and Wilderness Medicine
Oxford Handbook of Forensic Medicine
Oxford Handbook of Gastroenterology & Hepatology 2e
Oxford Handbook of General Practice 4e
Oxford Handbook of Genetics
Oxford Handbook of Genitourinary Medicine, HIV and AIDS 2e
Oxford Handbook of Geriatric Medicine 2e
Oxford Handbook of Infectious Diseases and Microbiology
Oxford Handbook of Key Clinical Evidence
Oxford Handbook of Medical Dermatology
Oxford Handbook of Medical Imaging
Oxford Handbook of Medical Sciences 2e
Oxford Handbook of Medical Statistics
Oxford Handbook of Neonatology
Oxford Handbook of Nephrology and Hypertension 2e
Oxford Handbook of Neurology 2e
Oxford Handbook of Nutrition and Dietetics 2e
Oxford Handbook of Obstetrics and Gynaecology 3e
Oxford Handbook of Occupational Health 2e
Oxford Handbook of Oncology 3e
Oxford Handbook of Ophthalmology 2e
Oxford Handbook of Oral and Maxillofacial Surgery
Oxford Handbook of Orthopaedics and Trauma
Oxford Handbook of Paediatrics 2e
Oxford Handbook of Pain Management
Oxford Handbook of Palliative Care 2e
Oxford Handbook of Practical Drug Therapy 2e
Oxford Handbook of Pre-Hospital Care
Oxford Handbook of Psychiatry 3e
Oxford Handbook of Public Health Practice 3e
Oxford Handbook of Reproductive Medicine & Family Planning 2e
Oxford Handbook of Respiratory Medicine 2e
Oxford Handbook of Rheumatology 3e
Oxford Handbook of Sport and Exercise Medicine 2e
Handbook of Surgical Consent
Oxford Handbook of Tropical Medicine 4e
Oxford Handbook of Urology 3e
Oxford Handbook of
Endocrinology
and Diabetes
Third edition
Edited by
John Wass
Professor of Endocrinology,
Oxford Centre for Diabetes,
Endocrinology and Metabolism (OCDEM),
Oxford, UK
Katharine Owen
Senior Clinical Researcher and Honorary Consultant,
Oxford Centre for Diabetes, Endocrinology and
Metabolism (OCDEM), Oxford, UK
Advisory editor
Helen Turner
Consultant in Endocrinology
Oxford Centre for Diabetes, Endocrinology and
Metabolism (OCDEM), Oxford, UK
1
3
Great Clarendon Street, Oxford, OX2 6DP,
United Kingdom
Oxford University Press is a department of the University of Oxford.
It furthers the University’s objective of excellence in research, scholarship,
and education by publishing worldwide. Oxford is a registered trade mark of
Oxford University Press in the UK and in certain other countries
© Oxford University Press 2014
The moral rights of the authors have been asserted
First edition published 2002
Second edition published 2009
Third edition published 2014
All rights reserved. No part of this publication may be reproduced,
stored in a retrieval system, or transmitted, in any form or by any means,
without the prior permission in writing of Oxford University Press,
or as expressly permitted by law, or under terms agreed with the appropriate
reprographics rights organization. Enquiries concerning reproduction
outside the scope of the above should be sent to the Rights Department,
Oxford University Press, at the address above
You must not circulate this work in any other form
and you must impose this same condition on any acquirer
Published in the United States of America by Oxford University Press
198 Madison Avenue, New York, NY 10016, United States of America
British Library Cataloguing in Publication Data
Data available
Library of Congress Control Number: 2013945298
ISBN 978–0–19–964443–8
Printed in China by
C&C Offset Printing Co. Ltd.
Oxford University Press makes no representation, express or implied, that the
drug dosages in this book are correct. Readers must therefore always check the
product information and clinical procedures with the most up-to-date published
product information and data sheets provided by the manufacturers and the most
recent codes of conduct and safety regulations. The authors and the publishers do
not accept responsibility or legal liability for any errors in the text or for the misuse
or misapplication of material in this work. Except where otherwise stated, drug
dosages and recommendations are for the non-pregnant adult who is not
breast-feeding.
Links to third party websites are provided by Oxford in good faith and
for information only. Oxford disclaims any responsibility for the materials
contained in any third party website referenced in this work.
v
Foreword
Philippe Bouchard
President, European Society of Endocrinology
Member of the National Academy of Medicine
vi
Preface to the
second edition
The first edition of this handbook was well received and sold many copies.
We were told by a number of specialist registrars in training and consult-
ants that it was essential to have it in outpatients. We hope that the same
will be true of the second edition.
Endocrinology remains the most exciting of specialties—enormously
varied in presentation and management and with the ability to affect hugely
and beneficially the quality of life over a long period of time. Our aims with
this second edition remain the same, mainly to have a pocket handbook
which can be easily transported in which all the pieces of information one
so often needs are there as a reminder. We hope it will enable trainees
to enhance their knowledge but also the older and so-called ‘trained’ will
continue to have recourse to its pages when memory lapses occur. We
regard it too as a companion to the Oxford Textbook of Endocrinology and
Diabetes.
We are enormously indebted to our contributors who once again have
provided timely texts full of practical detail. We are also hugely grateful to
our external referees who have looked at all the chapters with great care
and attention. Both have ensured that the text is as up-to-date as possible.
As always we welcome comments for future editions and we hope this
one proves as useful as the first one.
John A.H. Wass
Helen E.Turner
2009
vii
Preface
We remain happy that this handbook has been well received both in its
first and second editions. It has been translated into Chinese, and there
is also an American version which has sold well. We want it to remain
essential for specialist registrars in training and consultants who may have
the occasional memory lapse.
Our subject remains one of the most exciting of the specialties; our aims
with this third edition remain the same—to have, within a small volume,
all the essential information that one needs to look after patients with
endocrine problems and diabetes.
It is also an accompaniment to the Oxford Textbook of Endocrinology
and Diabetes which has recently been published in its second edition
(2011).
For this edition, we have completely revamped the diabetes section,
and we hope and think that this has been made more readily accessible
and assimilable.
We are enormously indebted to our contributors who have pro-
vided expertise and willing collaboration with our project. As always, we
welcome comments which may enhance the next edition.
Contents
Contributors x
Contributors to the second edition xii
Symbols and Abbreviations xiii
1 Thyroid 1
2 Pituitary 106
3 Adrenal 227
4 Reproductive endocrinology 297
5 Endocrinology in pregnancy 425
6 Calcium and bone metabolism 449
7 Paediatric endocrinology 513
8 Neuroendocrine disorders 553
9 Inherited endocrine syndromes and MEN 575
10 Endocrine surgery 601
11 Endocrinology and ageing 613
12 Endocrinology aspects of other clinical
or physiological situations 627
13 Diabetes 683
14 Lipids and hyperlipidaemia 823
15 Obesity 847
Appendix 1 867
Appendix 2 873
Appendix 3 881
Index 887
x
Contributors
Contributors to the
second edition
Julian Barth Peter Selby
Consultant in Chemical Pathology Consultant Physician and Senior
and Metabolic Medicine, Leeds Lecturer in Medicine, Manchester
General Infirmary, Leeds, UK Royal Infirmary, Manchester, UK
Karin Bradley Kevin Shotliff
Consultant Physician and Consultant Physician and
Endocrinologist, Bristol Royal Diabetologist, Beta Cell Diabetes
Infirmary and Honorary Senior Centre, Chelsea and Westminster
Clinical Lecturer, University of Hospital, London, UK
Bristol, Bristol, UK
Sara Suliman
Emma Duncan Specialist Registrar in Diabetes,
Consultant Endocrinologist, Endocrinology and Metabolism,
Princess Alexandra Hospital and Diabetes UK; Clinical
Senior Lecturer, University of Research Fellow, Oxford Centre
Queensland, Austalia; for Diabetes, Endocrinology and
Postdoctoral Research Fellow, UQ Metabolism (OCDEM), Churchill
Diamantina Institute for Cancer, Hospital, Oxford, UK
Immunology and Metabolic
Medicine, Australia Janet Sumner
Lead Diabetes Specialist Nurse,
Pam Dyson Churchill Hospital, Oxford, UK
Research Dietician, Oxford
University, Oxford, UK Vivien Thornton-Jones
Lead Endocrine Specialist Nurse,
Mohgah Elsheikh Churchill Hospital, Oxford, UK
Consultant Endocrinologist, Royal
Berkshire Hospital, Reading, UK Helen E. Turner
Consultant Endocrinologist,
Stephen Gardner Department of Endocrinology,
Consultant Physician, Churchill Hospital, Oxford, UK
Buckinghamshire Hospitals NHS
Trust, UK John Wong
Consultant Chemical Pathologist,
John Newell-Price Kingston Hospital, Surrey, UK
Senior Lecturer and Consultant
Endocrinologist, University of
Sheffield, Royal Hallamshire
Hospital, Sheffield, UK
xiii
Symbols and
Abbreviations
b cross-reference
7 approximately
i increased
d decreased
p primary
s secondary
α alpha
β beta
γ gamma
% per cent
♀ female
♂ male
+ve positive
–ve negative
= equal to
≡ equivalent to
< less than
> more than
≤ less than or equal to
≥ greater than or equal to
°C degree Celsius
£ pound Sterling
® registered trademark
2 important
3 don’t dawdle
ACA adrenocortical adenoma
ACC adrenocortical carcinoma
ACE angiotensin-converting enzyme
ACEI angiotensin-converting enzyme inhibitor
ACR albumin:creatinine ratio
ACTH adrenocorticotrophic hormone
AD autosomal dominant
ADA American Diabetes Association
ADH antidiuretic hormone
ADHH autosomal dominant hypocalcaemic hypercalciuria
xiv SYMBOLS AND ABBREVIATIONS
CF cystic fibrosis
CFRD CF-related diabetes
CGM continuous glucose monitoring
cGMP cyclic guanyl monophosphate
cGy centigray
CHD coronary heart disease
CHO carbohydrate
CK creatine kinase
CKD chronic kidney disease
CLAH congenital lipoid adrenal hyperplasia
cm centimetre
CMV cytomegalovirus
CNS central nervous system
COCP combined oral contraceptive pill
COPD chronic obstructive pulmonary disease
CPA cyproterone acetate
CPK creatine phosphokinase
Cr creatinine
CRF chronic renal failure
CRH corticotrophin-releasing hormone
CRP C-reactive protein
CSF cerebrospinal fluid
CSII continuous subcutaneous insulin infusion
CSMO ‘clinically significant’ diabetic macular oedema
CSW cerebral salt wasting
CT computed tomography
CTLA4 cytotoxic T lymphocyte antigen 4
cv coefficient of variation
CV cardiovascular
CVA cerebrovascular accident
CVD cardiovascular disease
CVP central venous pressure
CXR chest X-ray
DCCT Diabetes Control and Complications Trial
DCT distal convoluted tubule
DD disc diameter
DEXA dual-energy X-ray absorptiometry
DHEA dehydroepiandrostenedione
DHEAS dehydroepiandrostenedione sulphate
DHT dihydrotestosterone
xvi SYMBOLS AND ABBREVIATIONS
DI diabetes insipidus
DIT diiodotyrosine
DKA diabetic ketoacidosis
DKD diabetic kidney disease
dL decilitre
DM diabetes mellitus
DME diabetic macular (o)edema
DN diabetic neuropathy
DNA deoxyribonucleic acid
DOC deoxycorticosterone
DR diabetic retinopathy
DRIP/TRAP vitamin D receptor interacting protein/TR-associated
protein
DRS Diabetic Retinopathy Study
DSN diabetes specialist nurse
DSD disorders of sexual differentiation; disorders of sex
development
DTC differentiated thyroid cancer
DVA Driver and Vehicle Agency
DVLA Driver and Vehicle Licensing Agency
DVT deep vein thrombosis
DXA dual-energy absorptiometry
EBRT external beam radiation therapy
ECF extracellular fluid
ECG electrocardiogram
EEG electroencephalogram
eFPGL extra-adrenal functional paraganglioma
e.g. exempli gratia (for example)
eGFR estimated glomerular filtration rate
EM electron microscopy
EMA European Medicines Agency
ENaC epithelial sodium channel
ENETS European Neuroendocrine Tumour Society
ENSAT European Network for the Study of Adrenal Tumours
ENT ear, nose, and throat
EOSS Edmonton Obesity Staging System
ER (o)estrogen receptor
ERT (o)estrogen replacement therapy
ESR erythrocyte sedimentation rate
ESRD end-stage renal disease
ESRF end-stage renal failure
SYMBOLS AND ABBREVIATIONS xvii
kg kilogram
KPD ketosis-prone diabetes
L litre
LADA latent autoimmune diabetes of adulthood
LCAT lecithin:cholesterol acyltransferase
LDL low-density lipoprotein
LDL-C LDL cholesterol
LFT liver function test
LH luteinizing hormone
LOH loss of heterozygosity
Lpa lipoprotein a
LPL lipoprotein lipase
LVH left ventricular hypertrophy
m metre
MAI Mycobacterium avium intracellulare
MAOI monoamine oxidase inhibitor
MAPK mitogen-activated protein kinase
MBq mega becquerel
MC mineralocorticoid
MCR1 melanocortin 1 receptor
MDI multiple dose injection
MDT multidisciplinary team
MEN multiple endocrine neoplasia
mg milligram
Mg magnesium
MGMT O-6-methylguanine DNA methyltransferase
mGy milligray
MHC major histocompatibility complex
MI myocardial infarction
MIBG metaiodobenzylguanidine
min minute
MIS Müllerian inhibitory substance
MIT monoiodotyrosine
mIU milli international unit
MJ megajoule
mm millimetre
mmHg millimetre of mercury
MMI methimazole
mmol millimole
MODY maturity onset diabetes of the young
SYMBOLS AND ABBREVIATIONS xxi
mOsm milliosmole
MPH mid-parental height
MRI magnetic resonance imaging
mRNA messenger ribonucleic acid
MRSA meticillin-resistant Staphylococcus aureus
MSH melanocyte-stimulating hormone
MSU midstream urine
mSv microsievert
MTC medullary thyroid carcinoma
mTOR mammalian target of rapamycin
mU milliunit
Na sodium
NaCl sodium chloride
NAFLD non-alcoholic fatty liver disease
NASH non-alcoholic steatohepatitis
NaU urinary sodium
NB nota bene (take note)
NDST National Diabetes Support Team
NEC neuroendocrine carcinoma
NEN neuroendocrine neoplasia
NET neuroendocrine tumour
NF neurofibromatosis
NFA non-functioning pituitary adenoma
ng nanogram
NG nasogastric
NHS National Health Service
NICE National Institute for Health and Care Excellence
NIDDM non-insulin-dependent diabetes mellitus
NIS sodium/iodide symporter
nmol nanomole
NOGG National Osteoporosis Guideline Group
NR normal range
NSAID non-steroidal anti-inflammatory drug
NSC National Screening Committee
NSF National Service Framework
NVD new vessels on disc
NVE new vessels elsewhere
O2 oxygen
OA osteoarthritis
OCP oral contraceptive pill
xxii SYMBOLS AND ABBREVIATIONS
SU sulphonylurea
T3 tri-iodothyronine
T4 thyroxine
TART testicular adrenal rest tissue
TB tuberculosis
TBG thyroid-binding globulin
TBI traumatic brain injury
TBPA T4-binding prealbumin
TC total cholesterol
TCA tricyclic antidepressant
TDD total daily dose
T1DM type 1 diabetes mellitus
T2DM type 2 diabetes mellitus
tds ter die sumendus (three times daily)
TENS transcutaneous electrical nerve stimulation
TFT thyroid function test
Tg thyroglobulin
TG triglyceride
TGF transforming growth factor
TgAb thyroglobulin antibody
TK tyrosine kinase
TKI tyrosine kinase inhibitor
TNDM transient neonatal diabetes mellitus
TNF tumour necrosis factor
TPO thyroid peroxidase
TR thyroid hormone receptor
TRE thyroid hormone response element
TRH thyrotropin-releasing hormone
TSA transsphenoidal approach
TSAb TSH-stimulating antibody
TSG tumour suppressor gene
TSH thyroid-stimulating hormone
TSH-RAB thyroid-stimulating hormone receptor antibodies
TTR transthyretin
U unit
U&E urea and electrolytes
UFC urinary free cortisol
UK United Kingdom
UKPDS United Kingdom Prospective Diabetes Study
SYMBOLS AND ABBREVIATIONS xxv
US ultrasound
USA United States of America
V volts
VA visual acuity
VEGF vascular endothelial growth factor
VEGFR vascular endothelial growth factor receptor
VHL von Hippel–Lindau
VIP vasoactive intestinal polypeptide
VLCFA very long chain fatty acid
VLDL very low density lipoprotein
VMA vanillylmandelic acid
VRIII variable-rate intravenous insulin infusion
vs versus
VTE venous thromboembolism
WBS whole body scan
WDHA watery diarrhoea, hypokalaemia, acidosis
WHI Women’s Health Initiative
WHO World Health Organization
w/v weight by volume
ZE Zollinger–Ellison (syndrome)
Chapter 1 1
Thyroid
Anatomy 2
Physiology 4
Molecular action of thyroid hormone 6
Tests of hormone concentration 8
Tests of homeostatic control 10
Rare genetic disorders of thyroid hormone metabolism 14
Antibody screen 15
Scintiscanning 16
Ultrasound (US) scanning 18
Fine needle aspiration cytology (FNAC) 20
Computed tomography (CT) 22
Positron emission tomography (PET) 23
Additional laboratory investigations 24
Non-thyroidal illness 24
Atypical clinical situations 25
Thyrotoxicosis—aetiology 26
Manifestations of hyperthyroidism 28
Medical treatment 30
Radioiodine treatment 34
Surgery 38
Thyroid crisis (storm) 40
Subclinical hyperthyroidism 42
Thyrotoxicosis in pregnancy 44
Hyperthyroidism in children 48
Secondary hyperthyroidism 50
Graves’s ophthalmopathy 54
Medical treatment of Graves’s ophthalmopathy 58
Surgical treatment of Graves’s ophthalmopathy 60
Graves’s dermopathy 62
Thyroid acropachy 63
Multinodular goitre and solitary adenomas 64
Thyroiditis 68
Chronic autoimmune (atrophic or Hashimoto’s) thyroiditis 70
Other types of thyroiditis 72
Hypothyroidism 74
Subclinical hypothyroidism 78
Treatment of hypothyroidism 80
Congenital hypothyroidism 84
Amiodarone and thyroid function 86
Epidemiology of thyroid cancer 91
Aetiology of thyroid cancer 92
Papillary thyroid carcinoma 96
Follicular thyroid carcinoma (FTC) 99
Follow-up of papillary and FTC 100
Medullary thyroid carcinoma (MTC) 103
Anaplastic (undifferentiated) thyroid cancer 104
Lymphoma 105
2 CHAPTER 1 Thyroid
Anatomy
The thyroid gland comprises:
• A midline isthmus lying horizontally just below the cricoid cartilage.
• Two lateral lobes that extend upward over the lower half of the
thyroid cartilage.
The gland lies deep to the strap muscles of the neck, enclosed in the pre-
tracheal fascia, which anchors it to the trachea, so that the thyroid moves
up on swallowing.
Histology
• Fibrous septa divide the gland into pseudolobules.
• Pseudolobules are composed of vesicles called follicles or acini,
surrounded by a capillary network.
• The follicle walls are lined by cuboidal epithelium.
• The lumen is filled with a proteinaceous colloid, which contains the
unique protein thyroglobulin. The peptide sequences of T4 and T3 are
synthesized and stored as a component of thyroglobulin.
Development
• Develops from the endoderm of the floor of the pharynx with some
contribution from the lateral pharyngeal pouches.
• Descent of the midline thyroid precursor gives rise to the thyroglossal
duct, which extends from the foramen caecum near the base of the
tongue to the isthmus of the thyroid.
• During development, the posterior aspect of the thyroid becomes
associated with the parathyroid glands and the parafollicular C cells,
derived from the ultimo-branchial body (fourth pharyngeal pouch),
which become incorporated into its substance.
• The C cells are the source of calcitonin and give rise to medullary
thyroid carcinoma when they undergo malignant transformation.
• The fetal thyroid begins to concentrate and organify iodine at about
10–12 weeks’ gestation.
• Maternal TRH readily crosses the placenta; maternal TSH and T4
do not.
• T4 from the fetal thyroid is the major thyroid hormone available to the
fetus. The fetal pituitary-thyroid axis is a functional unit, distinct from
that of the mother—active at 18–20 weeks.
Thyroid examination
Inspection
• Look at the neck from the front. If a goitre (enlarged thyroid gland of
whatever cause) is present, the patient should be asked to swallow a
mouthful of water. The thyroid moves up with swallowing.
• Assess for scars, asymmetry, or masses.
• Watch for the appearance of any nodule not visible before swallowing;
beware that, in an elderly patient with kyphosis, the thyroid may be
partially retrosternal.
ANATOMY 3
Physiology
• Biosynthesis of thyroid hormones requires iodine as substrate.
Iodine is actively transported via sodium/iodide symporters (NIS)
into follicular thyrocytes where it is organified onto tyrosyl residues
in thyroglobulin first to produce monoiodotyrosine (MIT) and then
diiodotyrosine (DIT). Thyroid peroxidase (TPO) then links two DITs
to form the two-ringed structure T4, and MIT and DIT to form small
amounts of T3 and reverse T3 (rT3).
• The thyroid is the only source of T4.
• The thyroid secretes 20% of circulating T3; the remainder is generated
in extraglandular tissues by the conversion of T4 to T3 by deiodinases
(largely in the liver and kidneys).
Synthesis of the thyroid hormones can be inhibited by a variety of agents
termed goitrogens.
• Perchlorate and thiocyanate inhibit iodide transport.
• Thioureas (e.g. carbimazole and propylthiouracil) and mercaptoimidazole
inhibit the initial oxidation of iodide and coupling of iodothyronines.
• In large doses, iodine itself blocks organic binding and coupling
reactions.
• Lithium has several inhibitory effects on intrathyroidal iodine
metabolism.
In the blood, T4 and T3 are almost entirely bound to plasma proteins. T4 is
bound in d order of affinity to thyroid-binding globulin (TBG), transthyre-
tin (TTR), and albumin. T3 is bound 10–20 times less avidly by TBG and
not significantly by TTR. Only the free or unbound hormone is available to
tissues. The metabolic state correlates more closely with the free than the
total hormone concentration in the plasma. The relatively weak binding of
T3 accounts for its more rapid onset and offset of action. Table 1.1 sum-
marizes those states associated with p alterations in the concentration of
TBG. When there is primarily an alteration in the concentration of thyroid
hormones, the concentration of TBG changes little (Table 1.2).
The concentration of free hormones does not necessarily vary directly
with that of the total hormones, e.g. while the total T4 level rises in preg-
nancy, the free T4 level remains normal (b Endocrinology in pregnancy,
p. 426).
The levels of thyroid hormone in the blood are tightly controlled by
feedback mechanisms involved in the hypothalamo–pituitary–thyroid
(HPT) axis (see Fig. 1.1).
• TSH secreted by the pituitary stimulates the thyroid to secrete principally
T4 and also T3. TRH stimulates the synthesis and secretion of TSH.
• T4 and T3 are bound to TBG, TTR, and albumin. The remaining free
hormones inhibit the synthesis and release of TRH and TSH.
• T4 is converted peripherally to the metabolically active T3 or the
inactive rT3.
• T4 and T3 are metabolized in the liver by conjugation with glucuronate
and sulphate. Enzyme inducers, such as phenobarbital, carbamazepine,
and phenytoin, increase the metabolic clearance of the hormones
without d the proportion of free hormone in the blood.
PHYSIOLOGY 5
Hypothalamus
TRH
Other tissues
Anterior
pituitary
TSH
I
I T4 and T3 T4 and T3 + TBG TBG˙T4
TBG˙T3
Fig. 1.1 Regulation of thyroid function. Solid arrows indicate stimulation; broken
arrow indicates inhibitory influence. TRH, thyrotropin-releasing hormone; TSH,
thyroid-stimulating hormone; T4, thyroxine; T3, tri-iodothyronine; I, iodine; TBG,
thyroid-binding globulin.
6 CHAPTER 1 Thyroid
Abnormalities of development
• Remnants of the thyroglossal duct may be found in any position along
the course of the tract of its descent:
• In the tongue, it is referred to as ‘lingual thyroid’.
• Thyroglossal cysts may be visible as midline swellings in the neck.
• Thyroglossal fistula develops as an opening in the middle of
the neck.
• As thyroglossal nodules or
• The ‘pyramidal lobe’, a structure contiguous with the thyroid
isthmus which extends upwards.
• The gland can descend too far down to reach the anterior
mediastinum.
• Congenital hypothyroidism may result from failure of the thyroid
to develop (agenesis). More commonly, however, congenital
hypothyroidism reflects enzyme defects impairing hormone synthesis.
Further reading
Williams GR, Bassett JH (2011). Deiodinases: the balance of thyroid hormone: local control of
thyroid hormone action: role of type 2 deiodinase. J Endocrinol 209, 261–72.
8 CHAPTER 1 Thyroid
Reference
1. Gurnell M, Halsall DJ, Chatterjee VK (2011). What should be done when thyroid function tests
do not make sense? Clin Endocrinol (Oxf) 74, 673–8.
TESTS OF HOMEOSTATIC CONTROL 13
14 CHAPTER 1 Thyroid
1
Reviewed by Refetoff and Dumitrescu (2007) Syndromes of reduced sensitivity to thyroid hor-
mone: genetic defects in hormone receptors, cell transporters and deiodination. Best Pract Res Clin
Endocrinol Metab 21, 277–305.
ANTIBODY SCREEN 15
Antibody screen
High titres of antithyroid peroxidase (anti-TPO) antibodies and/or antithy-
roglobulin antibodies are found in patients with autoimmune thyroid dis-
ease (Hashimoto’s thyroiditis, Graves’s disease, and sometimes euthyroid
individuals). See Table 1.6.
Screening for thyroid disease1
The following categories of patients should be screened for thyroid
disease:
• Patients with atrial fibrillation or hyperlipidaemia.
• Periodic (6-monthly) assessments in patients receiving amiodarone and
lithium.
• Annual check of thyroid function in the annual review of diabetic
patients.
• ♀ with type 1 diabetes in the first trimester of pregnancy and
post-delivery (because of the 3-fold increase in incidence of
post-partum thyroid dysfunction in such patients) (b p. 432).
• ♀ with past history of post-partum thyroiditis.
• Annual check of thyroid function in people with Down’s syndrome,
Turner’s syndrome, and autoimmune Addison’s disease, in view of the
high prevalence of hypothyroidism in such patients.
• ♀ with thyroid autoantibodies—8x risk of developing hypothyroidism
over 20 years compared to antibody –ve controls.
• ♀ with thyroid autoantibodies and isolated elevated TSH—38x
risk of developing hypothyroidism, with 4% annual risk of overt
hypothyroidism.
• Maternal thyroid antibodies are associated with miscarriage and
preterm birth2.
Reference
1. Tunbridge WM, Vanderpump MP (2000). Population screening for autoimmune thyroid disease.
Endocrinol Metab Clin N Am 29, 239–53.
2. Thangaratinam S, et al. (2011). Association between thyroid autoantibodies and miscarriage and
preterm birth: meta-analysis of evidence. BMJ 342, 1065.
16 CHAPTER 1 Thyroid
Scintiscanning
Permits localization of sites of accumulation of radioiodine or sodium
pertechnetate (99mTc), which gives information about the activity of the
iodine trap (see Table 1.7). This is useful:
• To define areas of i or d function within the thyroid (see Table 1.8)
which occasionally helps in cases of uncertainty as to the cause of the
thyrotoxicosis.
• To distinguish between Graves’s disease and a thyroiditis (autoimmune
or viral—de Quervain’s thyroiditis).
• To detect retrosternal goitre.
• To detect ectopic thyroid tissue.
The scan may be altered by:
• Agents which influence thyroid uptake, including intake of high-iodine
foods and supplements, such as kelp (seaweed).
• Drugs containing iodine, such as amiodarone.
• Recent use of radiographic contrast dyes can potentially interfere with
the interpretation of the scan.
Thyrotoxicosis—aetiology
Epidemiology
• 10x more common in ♀ than in ♂ in the UK.
• Prevalence is approximately 2% of the ♀ population.
• Annual incidence is 3 cases per 1,000 ♀.
Definition of thyrotoxicosis and hyperthyroidism
• The term thyrotoxicosis denotes the clinical, physiological, and
biochemical findings that result when the tissues are exposed
to excess thyroid hormone. It can arise in a variety of ways (see
Table 1.11). It is essential to establish a specific diagnosis, as this
determines therapy choices and provides important information for
the patient regarding prognosis.
• The term hyperthyroidism should be used to denote only those
conditions in which hyperfunction of the thyroid leads to
thyrotoxicosis.
Genetics of autoimmune thyroid disease (AITD)
• AITD consists of Graves’s disease, Hashimoto’s thyroiditis,
atrophic autoimmune hypothyroidism, post-partum thyroiditis, and
thyroid-associated ophthalmopathy, that appear to share a common
genetic predisposition.
• There is a ♀ preponderance, and sex steroids appear to play an
important role.
• Twin studies show i concordance for Graves’s disease and
autoimmune hypothyroidism in monozygotic, compared to
dizygotic, twins.
• It is estimated that genetic factors account for approximately 70% of
the susceptibility for Graves’s disease.
• Sib studies indicate that sisters and children of ♀ with Graves’s disease
have a 5–8% risk of developing Graves’s disease or autoimmune
hypothyroidism.
• On the background of a genetic predisposition, environmental factors
are thought to contribute to the development of disease.
• A number of interacting susceptibility genes are thought to play a role
in the development of disease—a complex genetic trait.
• CTLA-4 (cytotoxic T lymphocyte antigen-4) is associated with Graves’s
disease in Caucasian populations. In particular, the CT60 allele has
a prevalence of 60% in the general population but is also the allele
most highly associated with Graves’s disease. These data emphasize
the complex nature of genetic susceptibility and the likely interplay of
environmental factors.
• Association of major histocompatibility complex (MHC) loci with
Graves’s disease has been demonstrated in some populations but
not others. HLA-DR3 is associated with Graves’s disease in whites.
HLA-DQA1*0501 is associated in some populations, especially for
men. However, the overall contribution of MHC genes to Graves’s
disease has been estimated to be only 10–20% of the inherited
susceptibility.
THYROTOXICOSIS—AETIOLOGY 27
Manifestations of hyperthyroidism
(See Box 1.2.)
Investigation of thyrotoxicosis
(See Table 1.12.)
• Thyroid function tests—raised free T4 and suppressed TSH (raised free
T3 in T3 toxicosis).
• TSH receptor antibodies—see b Table 1.6, p. 15. Also useful in the
assessment of cessation of carbimazole and in pregnant women to
assess the risk of fetal thyrotoxicosis.
• Radionucleotide thyroid scan if diagnosis uncertain (b see p. 17 but is
seldom required.
Manifestations of Graves’s disease
(in addition to those in Box 1.2)
• Diffuse goitre.
• Ophthalmopathy (b see Graves’s ophthalmopathy, p. 54).
• A feeling of grittiness and discomfort in the eye.
• Retrobulbar pressure or pain, eyelid lag or retraction.
• Periorbital oedema, chemosis,* scleral injection.*
• Exophthalmos (proptosis).*
• Extraocular muscle dysfunction.*
• Exposure keratitis.*
• Optic neuropathy.*
*
Combination of these suggests congestive ophthalmopathy. Urgent action necessary if: corneal
ulceration, congestive ophthalmopathy, or optic neuropathy (b see Graves’s ophthalmopathy, p. 54).
MANIFESTATIONS OF HYPERTHYROIDISM 29
Medical treatment
In general, the standard policy in Europe is to offer a course of antithyroid
drugs (ATD) first. In the USA, radioiodine is more likely to be offered as
first-line treatment.
Aims and principles of medical treatment
• To induce remission in Graves’s disease.
• Monitor for relapse off treatment, initially 6–8-weekly for 6 months,
then 6-monthly for 2 years, and then annually thereafter or sooner if
symptoms return.
• Use of a computerized thyroid follow-up register greatly facilitates
monitoring and reduces the necessity for outpatient appointments.
• For relapse, consider definitive treatment, such as radioiodine or
surgery. A second course of ATD almost never results in remission.
Choice of drugs—thionamides
• Carbimazole, which can be given as a once-daily dose, is usually the drug
of first choice in the UK. Carbimazole is converted to methimazole by
cleavage of a carboxyl side chain on first liver passage. It has a lower
rate of side effects when compared with PTU (14 vs 52%).
• Propylthiouracil (PTU) should never be used as a first-line agent in
either children or adults, with the possible exceptions of pregnant
women and patients with life-threatening thyrotoxicosis. PTU
use should be restricted to circumstances when neither surgery
nor radioactive iodine is a treatment option in a patient who has
developed a toxic reaction to carbimazole and antithyroid drug
therapy is needed.
• During the first trimester of pregnancy, propylthiouracil is the preferred
drug of choice because of the possible association of carbimazole with
aplasia cutis.
Action of thionamides
• Thyroid hormone synthesis is inhibited by blockade of the action of
thyroid peroxidase.
• Thionamides are especially actively accumulated in thyrotoxic tissue.
• Propylthiouracil also inhibits the deiodinase type 1 activity and
thus may have advantages when given at high doses in severe
thyrotoxicosis.
Dose and effectiveness
• 5mg of carbimazole is roughly equivalent to 50mg of propylthiouracil.
Propylthiouracil has a theoretical advantage of inhibiting the
conversion of T4 to T3, and T3 levels decline more rapidly after starting
the drug.
• 30–40% of patients treated with an ATD remain euthyroid 10 years
after discontinuation of therapy. If hyperthyroidism recurs after
treatment with an ATD, there is little chance that a second course
of treatment will result in permanent remission. Young patients,
smokers, those with large goitres, ophthalmopathy, or high serum
MEDICAL TREATMENT 31
Treatment regimen
Two alternative regimens are practised for Graves’s disease: dose titration
and block and replace.
Dose titration regime
• The p aim is to achieve a euthyroid state with relatively high drug
doses and then to maintain euthyroidism with a low stable dose. The
dose of carbimazole or propylthiouracil is titrated according to the
thyroid function tests performed every 4–8 weeks, aiming for a serum
free T4 in the normal range and a detectable TSH. High serum TSH
indicates the need for a dose reduction. TSH may remain suppressed
for some weeks after normalization of thyroid hormone levels.
• The typical starting dose of carbimazole is 20–30mg/day. Higher doses
(40–60mg) may be indicated in severe cases, with very high levels
of FT4.
• This regimen has a lower rate of side effects than the block and
replace regimen.
The treatment is continued for 18 months, as this appears to represent
the length of therapy which is generally optimal in producing the remission
rate of up to 40% at 5 years after discontinuing therapy.
• Relapses are most likely to occur within the first year and may be
more likely in the presence of a large goitre, ophthalmic Graves’s
disease, current smokers, and high T4 level at the time of diagnosis,
or the presence of TSH receptor antibodies at the end of treatment.
Men have a higher recurrence rate than women.
• Patients with multinodular goitres and thyrotoxicosis always relapse
on cessation of antithyroid medication, and definitive treatment
with radioiodine or surgery is usually advised. Long-term thionamide
therapy at low dose is also an option, particularly for the elderly.
Block and replace regimen
• After achieving a euthyroid state on carbimazole alone, carbimazole
at a dose of 40mg daily, together with T4 at a dose of 100 micrograms,
can be prescribed. This is usually continued for 6 months.
• The main advantages are fewer hospital visits for checks of thyroid
function and shorter duration of treatment.
• Most patients achieve a euthyroid state within 4–6 weeks of
carbimazole therapy.
• During treatment, FT4 values are measured 4 weeks after starting
levothyroxine and the dose of levothyroxine altered, if necessary, in
25 micrograms increments to maintain FT4 in the normal range.
Most patients do not require any dose adjustment.
• The originally reported higher remission rate was not confirmed in
a large prospective multicentre European trial when combination
treatment was compared to carbimazole alone, but side effects were
more common.1
• Relapses are most likely to occur within the first year.
MEDICAL TREATMENT 33
Reference
1. Reinwein D, Benker G, Lazarus JH, et al. (1993). A prospective randomized trial of antithyroid
drug dose in Graves’s disease therapy. European Multicenter Study Group on Antithyroid Drug
Treatment. J Clin Endocrinol Metab 76, 1516–21.
34 CHAPTER 1 Thyroid
Radioiodine treatment
(See Table 1.13.)
Indications
• Definitive treatment of multinodular goitre or adenoma.
• Relapsed Graves’s disease.
Contraindications
• Young children because of the potential risk of thyroid carcinogenesis.
• Pregnant and lactating ♀.
• Situations where it is clear that the safety of other people cannot be
guaranteed.
• Graves’s ophthalmopathy. There is some evidence that Graves’s
ophthalmopathy may worsen after the administration of radioactive
iodine, especially in smokers. In cases of moderate-to-severe
ophthalmopathy, radioiodine may be avoided. Alternatively, steroid
cover in a dose of 40mg prednisolone should be administered on
the day of administering radioiodine, 30mg daily for the next
2 weeks, 20mg daily for the following 2 weeks, reducing to zero over
subsequent 3 weeks. Lower steroid doses, e.g. starting with 20mg,
may be effective. It is essential that euthyroidism is closely maintained
following radioiodine to avoid worsening of ophthalmopathy.
Caveats
• The control of disease may not occur for a period of weeks or a few
months.
• More than one treatment may be needed in some patients, depending
on the dose given; 15% require a second dose, and a few patients
require a third dose. The second dose should be considered only at
least 6 months after the first dose.
• Compounds that contain iodine, such as amiodarone, block iodine
uptake for a period of several months following cessation of therapy;
iodine uptake measurements may be helpful in this instance in
determining the activity required and the timing of radioiodine therapy.
• ♀ of childbearing age should avoid pregnancy for a minimum of
6 months following radioactive iodine ablation.
• Men should avoid fathering children for 4 months after radiation.
• The prevalence of hypothyroidism is about 50% at 10 years and
continues to increase thereafter.
Side effects are rare
• Anterior neck pain caused by radiation-induced thyroiditis (1%).
• Transient rise (72h) in thyroid hormone levels which may exacerbate
heart failure, if present. This aspect needs consideration in elderly
patients.
RADIOIODINE TREATMENT 35
Reference
1. Royal College of Physicians of London (2007). The use of radioiodine in benign thyroid disease.
Royal College of Physicians, London.
38 CHAPTER 1 Thyroid
Surgery
(Aspects of thyroid surgery are also covered b see p. 606.)
Total thyroidectomy is now considered the operation of choice because
of the risk of relapse with partial thyroidectomy. All such patients go home
on T4. See Box 1.3.
Indications
• Documented suspicious or malignant thyroid nodule by FNAC.
• Pregnant women who are not adequately controlled by ATDs or in
whom serious allergic reactions develop while being treated medically.
Thyroidectomy is usually performed in the second trimester.
• Patients:
• Who reject or fear exposure to radiation.
• With poor compliance to medical treatment.
• In whom a rapid control of symptoms is desired.
• With severe manifestations of Graves’s ophthalmopathy, as total or
near total thyroidectomy does not worsen eye manifestations (but
carefully avoid post-operative hypothyroidism).
• With relapsed Graves’s disease.
• With local compressive symptoms which may not improve rapidly
with radioiodine whereas operation removes these symptoms in
most patients.
• With large thyroid glands and relatively low radioiodine uptake.
Preparation of patients for surgery
• ATDs should be used preoperatively to achieve euthyroidism.
• Propranolol may be added to achieve B-blockade, especially in those
patients where surgery must be performed sooner than achieving
euthyroid state.
• Potassium iodide, 60mg 3x daily, can be used during the preoperative
period to prevent an unwanted liberation of thyroid hormones during
surgery. Preoperatively, it should be given for 10 days. Operating later
than this can be associated with exacerbation of thyrotoxicosis, as the
thyroid escapes from the inhibitory effect of the iodide. In practice, it is
rarely needed, as good control of thyrotoxicosis can be achieved with
ATDs in the majority of patients.
• In the patient who appears to be non-compliant with ATDs and
remains thyrotoxic prior to surgery, it may be necessary to admit
them as an inpatient for supervised administration of high-dose ATDs,
together with B-blockade, and measurement of FT4 and FT3 twice
weekly. There is a risk of thyroid crisis or storm if a patient undergoes
operation when thyrotoxic. Most patients can be rendered euthyroid
within 2–4 weeks, and potassium iodide can be administered as above
in this section to coincide with the timing of surgery.
• Additional measures are as for thyroid storm.
SURGERY 39
Specific treatment
• Aim: to inhibit thyroid hormone synthesis completely.
• Propylthiouracil 200–300mg 6-hourly via NG tube. Propylthiouracil
is preferred because of its ability to block T4 to T3 conversion
in peripheral tissues. There are no clinical data comparing
propylthiouracil and carbimazole in this situation. ATDs should be
commenced first.
• Potassium iodide, 60mg via NG tube 6-hourly, 6h after starting
propylthiouracil, will inhibit thyroid hormone release.
• B-adrenergic blocking agents are essential in the management to
control tachycardia, tremor, and other adrenergic manifestations:
• Propranolol 160–480mg/day in divided doses or as an infusion at a
rate of 2–5mg/h.
• Calcium channel blockers can be tried in patients with known
bronchospastic disease where B-blockade is contraindicated.
• High doses of glucocorticoids are capable of blocking T4 to T3
conversion: prednisolone 60mg daily or hydrocortisone 400mg IM,
4x daily.
• Plasmapheresis and peritoneal dialysis may be effective in cases
resistant to the usual pharmacological measures.
• Colestyramine (3g tds) reduces the enterohepatic circulation of
thyroid hormones and may help improve thyrotoxicosis.
42 CHAPTER 1 Thyroid
Subclinical hyperthyroidism
(See Box 1.5.)
• Values of thyroid hormones should be repeated to exclude
non-thyroidal illness.
• Subclinical hyperthyroidism is defined as low serum thyrotropin (TSH)
concentration in patients with normal levels of T4 and T3. Subtle
symptoms and signs of thyrotoxicosis may be present.
• May be classified as endogenous in patients with thyroid hormone
production associated with nodular thyroid disease or underlying
Graves’s disease; and as exogenous in those with low or undetectable
serum thyrotropin concentrations as a result of treatment with
levothyroxine.
There is epidemiological evidence that subclinical hyperthyroidism is
a risk factor for the development of atrial fibrillation or osteoporosis.1
Meta-analyses suggest a 41% increase in all-cause mortality.2
Further reading
Rhee EP, et al. (2012). Case 4-2012—A 37-year-old man with muscle pain, weakness, and weight
loss. New Engl J Med 366, 553.
44 CHAPTER 1 Thyroid
Thyrotoxicosis in pregnancy
(b also see p. 428 and Box 1.6.)
• Thyrotoxicosis occurs in about 0.2% of pregnancies.
• Graves’s disease accounts for 90% of cases.
• Less common causes include toxic adenoma and multinodular goitre.
• Other causes are gestational hyperthyroidism (hyperemesis
gravidarum) and trophoblastic neoplasia.
• Diagnosis of thyrotoxicosis during pregnancy may be difficult or
delayed.
• Physiological changes of pregnancy are similar to those of
hyperthyroidism.
• Total T4 and T3 are elevated in pregnancy because of an elevated level
of TBG, but, with free hormone assays available, this is no longer a
problem.
• Physiological features of normal pregnancy include an increase in
basal metabolic rate, cardiac stroke volume, palpitations, and heat
intolerance.
• Serum free T3 concentrations remain within the normal range in most
pregnant ♀; serum TSH concentration decreases during the first
trimester.
Symptoms
• Hyperemesis gravidarum is the classic presentation (one-third is toxic).
Tiredness, palpitations, insomnia, heat intolerance, proximal muscle
weakness, shortness of breath, and irritability may be other presenting
symptoms.
• Thyrotoxicosis may occasionally be diagnosed when the patient
presents with pregnancy-induced hypertension or congestive heart
failure.
Signs
• Failure to gain weight despite a good appetite.
• Persistent tachycardia with a pulse rate >90 beats/min at rest.
• Other signs of thyrotoxicosis as described previously.
Natural history of Graves’s disease in pregnancy
There is aggravation of symptoms in the first half of the pregnancy; amelio-
ration of symptoms in the second half of the pregnancy, and often recur-
rence of symptoms in the post-partum period.
Transient hyperthyroidism of gestational
hyperthyroidism (hyperemesis gravidarum)
• The likely mechanism is a raised B-hCG level.
• B-hCG, LH, FSH, and TSH are glycoprotein hormones that contain
a common α subunit and a hormone-specific B subunit. There is an
inverse relationship between the serum levels of TSH and hCG, best
seen in early pregnancy. There is also structural homology of the TSH
and hCG receptors.
THYROTOXICOSIS IN PREGNANCY 45
Hyperthyroidism in children
Epidemiology
Thyrotoxicosis is rare before the age of 5 years. Although there is a pro-
gressive increase in incidence throughout childhood, it is still rare and
accounts for <5% of all cases of Graves’s disease.
Clinical features
• Behavioural abnormalities, hyperactivity, declining school performance
may bring the child to medical attention. Features of hyperthyroidism
are as described previously.
• Acceleration of linear growth is common in patients increasing in
height percentiles on the growth charts. The disease may be part of
McCune–Albright syndrome, and café-au-lait pigmentation, precocious
puberty, and bony abnormalities should be considered during clinical
examination.
Investigations
The cause of thyrotoxicosis in children is nearly always Graves’s disease
(with +ve TSH receptor antibodies), although thyroiditis and toxic nod-
ules have been described, and a radioiodine scan may be useful if the
diagnosis is not clear. Hereditary syndromes of thyroid hormone resist-
ance, often misdiagnosed as Graves’s disease, are now being increasingly
recognized in children.
Treatment
ATDs represent the initial treatment of choice for thyrotoxic children.
Therapy is generally started with carbimazole 250 micrograms/kg (ini-
tial dose 10mg/day). Since relapse after withdrawal of ATDs is com-
mon, definitive treatment with surgery or radioiodine should be offered.
Hepatotoxicity with propylthiouracil can also occur in children.
HYPERTHYROIDISM IN CHILDREN 49
50 CHAPTER 1 Thyroid
Secondary hyperthyroidism
An elevated serum free T4 and non-suppressed serum TSH are character-
istic of TSH-secreting adenomas or resistance to thyroid hormone.
These conditions must be differentiated (see Table 1.15).
TSH-secreting pituitary tumours
• Estimated incidence 1 per million and <1% of all pituitary tumours.
• There are characteristically elevated serum free T4 and T3
concentrations and non-suppressed (inappropriately normal or frankly
elevated) serum TSH levels.
• Approximately 25% of TSHomas co-secrete one or more other
pituitary hormones; about 15% secrete growth hormone, 10%
prolactin, and rarely gonadotrophins. Approximately 90% are
macroadenomas (>1cm in diameter), and over two-thirds exhibit
suprasellar extension, invasion, or both into adjacent tissues (b see
p. 180).
• Patients with pure TSHomas present with typical symptoms and signs
of thyrotoxicosis and the presence of a diffuse goitre. Patients may
exhibit features of oversecretion of the other pituitary hormones,
e.g. prolactin or growth hormone. Headaches, visual field defects,
menstrual irregularities, amenorrhoea, delayed puberty, and
hypogonadotrophic hypogonadism have also been reported. Careful
establishment of the diagnosis is the key to treatment. Inappropriate
treatment of such patients with subtotal thyroidectomy or radioiodine
administration not only fails to cure the underlying disorder, but may
be associated with subsequent pituitary tumour enlargement and an i
risk of invasiveness into adjacent tissues.
• Treatment options are:
• Transphenoidal surgery.
• Pituitary radiotherapy if surgical results are unsatisfactory or surgery
is contraindicated or not desired.
• Medical therapy with somatostatin analogues, such as octreotide
or lanreotide, may be useful preoperatively and suppresses TSH
secretion in 80% of the cases.
Resistance to thyroid hormones
• Patients with generalized resistance to thyroid hormone (GRTH)
may present with mild hyperthyroidism, deaf mutism, delayed bone
maturation, raised circulating thyroid hormone concentrations,
non-suppressed TSH, and failure of TSH to decrease normally upon
administration of supraphysiological doses of thyroid hormones. Most
patients present with goitre or incidentally found abnormal TFTs.
Treatment is determined by thyroid status.
• The estimated incidence is 1:50,000 live births.
• In selective pituitary resistance to thyroid hormones (PRTH), the
thyroid hormone resistance is more pronounced in the pituitary; thus,
the patient exhibits definite clinical manifestations of thyrotoxicosis.
SECONDARY HYPERTHYROIDISM 51
Further reading
Abalovich M, Amino N, Barbour LA, et al. (2007). Management of thyroid dysfunction during preg-
nancy and postpartum: an Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab
92(8 Suppl), S1–47.
Abraham P, Avenell A, McGeoch SC, et al. (2010). Antithyroid drug regimen for treating Graves’s
hyperthyroidism. Cochrane Database Syst Rev CD003420.
Bahn RS, Burch HB, Cooper DS, et al. (2011). Hyperthyroidism and other causes of thyrotoxico-
sis: management guidelines of the American Thyroid Association and American Association of
Clinical Endocrinologists. Thyroid 21, 593–646.
Bauer AJ (2011). Approach to the pediatric patient with Graves’s disease: when is definitive therapy
warranted? J Clin Endocrinol Metab 96, 580–8.
Bonnema SJ, Hegedüs L (2012). Radioiodine therapy in benign thyroid diseases. Endocr Rev 33,
920–80.
Brand OJ, Gough SC (2010). Genetics of thyroid autoimmunity and the role of the TSHR. Mol Cell
Endocrinol 322, 135–43.
Brent GP (2008). Graves’s disease. N Engl J Med 358, 2594–605.
Brent GA (2010). Environmental exposures and autoimmune thyroid disease. Thyroid 20, 755–61.
Brix TH, Hegedüs L (2011). Twins as a tool for evaluating the influence of genetic susceptibility in
thyroid autoimmunity. Ann Endocrinol (Paris) 72, 103–7.
Cesur M, Bayram F, Temel MA, et al. (2008). Thyrotoxic hypokalaemic periodic paralysis in a
Turkish population: three new case reports and analysis of the case series. Clin Endocrinol (Oxf)
68, 143–152.
Clementi M, Di Gianantonio E, Cassina M, et al. (2010). Treatment of hyperthyroidism in pregnancy
and birth defects. J Clin Endocrinol Metab 95, E337–41.
Cooper DS, Rivkees SA (2009). Putting propylthiouracil in perspective. J Clin Endocrinol Metab
94, 1881–2.
Franklyn JA (2009). Thyroid gland: Antithyroid therapy--best choice of drug and dose. Nat Rev
Endocrinol 5, 592–4.
Franklyn JA (2010). What is the role of radioiodine uptake measurement and thyroid scintigraphy in
the diagnosis and management of hyperthyroidism? Clin Endocrinol (Oxf) 72, 11–12
Metso S, Jaatinen P, Huhtala H, et al. (2007). Increased cardiovascular and cancer mortality after
radioiodine treatment for hyperthyroidism. J Clin Endocrinol Metab 92, 2190–6.
Pearce SH (2004). Spontaneous reporting of adverse reactions to carbimazole and propylthiouracil
in the UK. Clin Endocrinol 61, 589–94.
Ross DS (2011). Radioiodine therapy for hyperthyroidism. N Engl J Med 364, 542–50.
Vanderpump MP (2011). Should we treat mild subclinical/mild hyperthyroidism? No. Eur J Intern
Med 22, 330–3.
Wiersinga WM (2011). Should we treat mild subclinical/mild hyperthyroidism? Yes. Eur J Intern
Med 22, 324–9.
SECONDARY HYPERTHYROIDISM 53
54 CHAPTER 1 Thyroid
Graves’s ophthalmopathy
(See European Working Group on Graves’s Ophthalmopathy,
M http://www.EUGOGO.org.)
• An organ-specific autoimmune disorder characterized by swelling of
the extraocular muscles, lymphocytic infiltration, late fibrosis, muscle
tethering, and proliferation of orbital fat and connective tissue.
• The volume of both the extraocular muscles and retroorbital
connective and adipose tissue is increased due to inflammation and
the accumulation of hydrophilic glycosaminoglycans (GAG), principally
hyaluronic acid, in these tissues. GAG secretion by fibroblasts is
increased by activated T-cell cytokines, such as tumour necrosis factor
(TNF) alpha and interferon gamma, implying that T-cell activation is an
important part of this immunopathology. The accumulation of GAG
causes a change in osmotic pressure, which, in turn, leads to a fluid
accumulation and an increase in pressure within the orbit. These changes
displace the eyeball forward and can also interfere with the function of
the extraocular muscles and the venous drainage of the orbits.
• Clinically evident in 720% of patients, but a further 730% may have
evidence on imaging. Most bilateral, but often asymmetrical, and 15%
have unilateral disease.
• Most have mild self-limiting disease, but 75% (more ♂ and the
elderly) have severe disease that threatens sight. The natural history is
variable, with spontaneous amelioration in 66%, no change in 20%, and
worsening in 14%.
• Incidence higher in ♀ (except for severe disease where equal sex
incidence). Prevalence decreasing (? associated with decreased
smoking).
• Bimodal age distribution in ♀, with peak onsets between 40–44 years
and 60–64 years. In ♂, a single peak incidence occurs at 65–69 years.
• There are two stages in the development of the disease, which can be
recognized as an active inflammatory (dynamic) stage and a relatively
quiescent static stage.
• The appearance of eye disease follows a different time course to
thyroid dysfunction, and, in a minority, there is a lag period between
the presentation of hyperthyroidism and the appearance of eye signs.
85% of patients develop Graves’s disease within 18 months of Graves’s
ophthalmopathy developing (20% precede, 40% following).
• 5% of patients with Graves’s ophthalmopathy have hypothyroidism,
and 5% are euthyroid.
• High levels of TSH receptor antibodies identify high-risk patients.
• Current smokers (>20/day) are more likely to develop ophthalmopathy.
• Continuing smoking and uncontrolled hyperthyroidism or
hypothyroidism moderately worsen Graves’s ophthalmopathy.
• The role of an endocrinologist during a routine review of Graves’s
patients is to record accurately the clinical features of Graves’s
eye disease and to identify ocular emergencies, such as corneal
ulceration, congestive ophthalmopathy, and optic neuropathy, which
should be referred urgently to an ophthalmologist, preferably in a
multidisciplinary clinic setting.
GRAVES’S OPHTHALMOPATHY 55
Clinical features
(See Box 1.7.)
• Retraction of eyelids is extremely common in thyroid eye disease.
The margin of the upper eyelid normally rests about 2mm below the
limbus, and retraction can be suspected if the lid margin is either level
with or above the superior limbus, allowing the sclera to be visible.
The lower lid normally rests at the inferior limbus, and retraction is
suspected when the sclera shows above the lid.
• Proptosis or exophthalmos can result in failure of lid closure,
increasing the likelihood of exposure keratitis and the common
symptom of gritty eyes. This can be confirmed with fluorescein or
Rose Bengal stain. As papilloedema can occur, fundoscopy should be
performed. Proptosis may result in periorbital oedema and chemosis
because the displaced orbit results in less efficient orbital drainage.
• Persistent visual blurring may indicate an optic neuropathy and
requires urgent treatment.
• Severe conjunctival pain may indicate corneal ulceration, requiring
urgent referral.
Investigation of proptosis
For details of the ‘NOSPECS’ classification, see M http://www.EUGOGO.
org and The European Group on Graves’s Orbitopathy. However,
NOSPECS is not always satisfactory for prospective objective assessment
of orbital changes, and determining an overall activity score is sometimes
more helpful. This is done by assigning one point for the presence of
each of the following findings: spontaneous retrobulbar pain, pain on eye
movement, eyelid erythema, conjunctival injection, chemosis, swelling of
the carbuncle, and eyelid oedema.
Orbital radiotherapy
• Indications for lens-sparing orbital radiotherapy are similar to those for
high-dose glucocorticoids.
• Radiotherapy works by killing retroorbital T cells.
• 20 Gray delivered over ten fractions is the standard regimen.
• Treatment with both radiotherapy and glucocorticoids is more
effective than either alone.
• Effectiveness in 60% of cases <40 years.
Other medical therapies
• Selenium1 may improve symptoms in patients with mild Graves’s
ophthalmopathy, as illustrated by the results of a randomized trial of
selenium (100 micrograms bd), pentoxifylline (600mg bd), or placebo
in 159 patients. After 6 months of treatment, eyelid aperture (37%
vs 12%) and soft tissue signs (43% vs 32%) significantly improved in
patients taking selenium vs placebo. Compared with placebo, selenium
also significantly improved quality of life (both visual functioning and
appearance scores), as assessed by the Graves’s Ophthalmopathy
Quality of Life Questionnaire (GO-QOL). Evaluation at 12 months
confirmed the findings at 6 months.
• A number of reports have indicated that some patients with severe
Graves’s ophthalmopathy may respond to B cell depletion induced by
rituximab, which is a monoclonal antibody directed against the B cell
CD20 molecule. Rituximab induces a fall in TSH receptor antibody
levels and depletion of B cells in the retroorbital tissues, not just the
periphery. Although high doses of this antibody may be associated with
severe side effects from profound immunosuppression, it is possible
that much lower doses may be effective in Graves’s ophthalmopathy
to avoid such effects. This approach to the treatment of severe eye
disease is currently undergoing larger trials; preliminary results from
these trials suggest efficacy in some, but not all, patients.
• Other immunosuppressive regimens have no proven place in the
general management of Graves’s ophthalmopathy.
• Use of depot octreotide has been shown to be of no benefit in
management.
Reference
1. Marcocci C, et al. (2011). Selenium and the course of mild Graves’ orbitopathy. N Engl J Med
364, 1920.
60 CHAPTER 1 Thyroid
Graves’s dermopathy
• This is a rare complication of Graves’s thyrotoxicosis (0.5%). It
is usually pretibial in location (99%) and hence called pretibial
myxoedema.
• Associated with ophthalmopathy (97%) and acropachy (18%).
• It typically appears as raised, discoloured, and indurated lesions on
the front or back of the legs or on the dorsum of the feet and has
occasionally been described in other areas, including the hands and
the face.
• The lesions are due to localized accumulation of glycosaminoglycans.
It is now recognized that there is a lymphocytic infiltrate. Lesions
are characteristically asymptomatic, but they can also be pruritic and
tender. They can be very disfiguring.
• Treatment. Usually not treated. Potent topical fluorinated steroids,
such as fluocinolone acetonide, may be effective (4–8 weeks), not only
in the treatment of localized pain and tenderness, but also in some
resolution of the visible skin signs. Surgery may worsen the condition.
• 25% remit completely; 50% are chronic on no therapy. A beneficial
effect of topical steroids on remission rates is unproven.
THYROID ACROPACHY 63
Thyroid acropachy
• This is the rarest manifestation of Graves’s disease.
• It presents as clubbing of the digits and subperiosteal new bone
formation. The soft tissue swelling is similar to that seen in localized
myxoedema and consists of glycosaminoglycan accumulation.
• Patients almost inevitably have Graves’s ophthalmopathy or pretibial
myxoedema. If not, an alternative cause of clubbing should be
looked for.
• It is typically painless, and there is no effective treatment.
Further reading
Bahn RS (2010). Graves’s ophthalmopathy. N Engl J Med. 362, 726–38.
Banga JP, Nielsen CH, Gilbert JA, et al. (2008). Application of new therapies in Graves’s disease
and thyroid-associated ophthalmopathy: animal models and translation to human clinical trials.
Thyroid 18, 973–81.
Bartalena L (2010). What to do for moderate-to-severe and active Graves’s orbitopathy if gluco-
corticoids fail? Clin Endocrinol (Oxf). 73, 149–52.
European Group on Graves’s orbitopathy (EUGOGO) (2008). Consensus statement of the
European Group on Graves’s orbitopathy (EUGOGO) on management of GO. Eur J Endocrinol.
158, 273–85.
Hegedüs L, Smith TJ, Douglas RS, et al. (2011). Targeted biological therapies for Graves’s dis-
ease and thyroid-associated ophthalmopathy. Focus on B-cell depletion with Rituximab. Clin
Endocrinol (Oxf). 74, 1–8.
Marcocci C, Kahaly GJ, Krassas GE, et al. (2011). Selenium and the course of mild Graves’s orbit-
opathy. N Engl J Med. 364, 1920–31.
Schwartz KM, Fatourechi V, Ahmed DD, et al. (1992). Dermopathy of Graves’s disease (pretibial
myxedema): long-term outcome. J Clin Endocrinol Metab 87, 438–46.
Stiebel-Kalish H, Robenshtok E, Hasanreisoglu M, et al. (2009). Treatment modalities for Graves’s
ophthalmopathy: systematic review and metaanalysis. J Clin Endocrinol Metab. 94, 2708–16.
Träisk F, Tallstedt L, Abraham-Nordling M, et al. (2009). Thyroid-associated ophthalmopathy after
treatment for Graves’s hyperthyroidism with antithyroid drugs or iodine-131. J Clin Endocrinol
Metab. 94, 3700–7.
The European Group on Graves’s Orbitopathy (2006). Clinical assessment of patients with
Graves’s orbitopathy: recommendations to generalists, specialists and clinical researchers. Eur
J Endocrinol 155, 387–9.
Zang S, Ponto KA, Kahaly GJ (2011). Clinical review: Intravenous glucocorticoids for Graves’s
orbitopathy: efficacy and morbidity. J Clin Endocrinol Metab. 96, 320–32.
64 CHAPTER 1 Thyroid
Radioiodine
• This form of treatment is often considered as first choice for definitive
treatment. 131I is preferentially accumulated in hot nodules but not
in normal thyroid tissue which, because of the thyrotoxic state, is
non-functioning.
• Radioiodine treatment commonly induces a euthyroid state, as
the hot nodules are destroyed and the previously non-functioning
follicles gradually resume normal function. A dose of 500–800MBq for
small-to-medium and 600 or 800MBq for medium-to-large goitres is
recommended.
Surgery
• The aim of surgery is to remove as much of the nodular tissue
as possible and, if the goitre is large, to relieve local symptoms.
Post-operative follow-up should involve checks of thyroid function.
• Goitre recurrence, although rare, does occasionally occur.
Non-toxic multinodular goitre
Surgery
• Is the preferred treatment for patients with:
• Local compression symptoms.
• Cosmetic disfigurement.
• Solitary nodule with FNAC suspicious of malignancy.
Radioiodine
• Radioiodine may be particularly indicated in elderly patients in whom
surgery is not appropriate. It may require admission. Up to 50%
shrinkage of goitre mass has been reported in recent studies.
• Hypothyroidism following radioiodine is relatively infrequent but is
still recognized.
Medical treatment
Use of T4 to suppress TSH is associated with risk of cardiac arrhythmias
and bone loss. T4 is useful only if TSH is detectable but is not generally
indicated.
Pathology
• Thyroid nodules may be described as adenomas if the follicular cell
differentiation is enclosed within a capsule; adenomatous when the
lesions are circumscribed but not encapsulated.
• The most common benign thyroid tumours are the nodules of
multinodular goitres (colloid nodules) and follicular adenomas. The
oncogene changes accounting for these benign thyroid nodules are
not well delineated. Multinodular goitres are occasionally familial,
which means that the patient has at least one germline mutation. One
familial form of non-toxic multinodular goitre has been linked to DNA
markers on chromosome 14q, but the aetiologic gene is not known.
Follicular adenomas are clonal, and approximately 25% of sporadic
follicular adenomas have a hemizygous deletion of a chromosome
region containing PTEN (MMAC1), the tumour suppressor gene in
which germline defects cause Cowden syndrome (see b p. 584).
• Autonomously functioning thyroid adenomas (or nodules) are benign
tumours that produce thyroid hormone. Clinically, they present as a
single nodule that is hyperfunctioning (‘hot’) on thyroid radionuclide
scan, sometimes causing hyperthyroidism. Many of these tumours are
caused by somatic mutations in genes that code for the TSH receptor
and α subunit of the guanyl nucleotide stimulatory protein (Gs).
• Activating mutations of the TSH receptor produce constitutive
activation of adenylyl cyclase in the absence of TSH. The thyroid
follicular cell with this TSH receptor mutation divides and produces
thyroid hormone without TSH stimulation, eventually becoming
clinically recognized as a hot nodule. Among patients with an
autonomously functioning thyroid adenoma, the frequency of TSH
receptor mutations in the adenoma varies from 75% to 80%. Since
the mutations are scattered throughout the receptor, studies of
the entire receptor are most likely to identify a mutation. In rare
families, germline mutations in the TSH receptor cause hereditary
hyperthyroidism, initially with a diffuse goitre but ultimately with a
nodular goitre with multiple hot nodules.
Thyroid nodules in pregnant women
• Increase in size during gestation.
• Increase in number.
• Need FNA as higher risk of malignancy.
• Can be operated upon in second trimester or post-partum.
Further reading
Bahn RS, Castro MR (2011). Approach to the patient with nontoxic multinodular goiter. J Clin
Endocrinol Metab 96, 1202–12.
Boelaert K (2009). The association between serum TSH concentration and thyroid cancer. Endocr
Relat Cancer 16, 1065–72.
Eszlinger M, Jaeschke H, Paschke R (2007). Insights from molecular pathways: potential pharmaco-
logic targets of benign thyroid nodules. Curr Opin Endocrinol Diabetes Obes 14, 393–7.
Fast S, Bonnema SJ, Hegedüs L (2011). Radioiodine therapy of benign non-toxic goitre. Potential
role of recombinant human TSH. Ann Endocrinol (Paris) 72, 129–35.
Lueblinghoff J, Eszlinger M, Jaeschke H, et al. (2011). Shared sporadic and somatic thyrotropin
receptor mutations display more active in vitro activities than familial thyrotropin receptor
mutations. Thyroid 21, 221–9.
68 CHAPTER 1 Thyroid
Thyroiditis
Background
Inflammation of the thyroid gland often leads to a transient thyrotoxicosis
followed by hypothyroidism. Overt hypothyroidism caused by autoim-
munity has two main forms: Hashimoto’s (goitrous) thyroiditis and atrophic
thyroiditis. See Tables 1.16 and 1.17.
Hypothyroidism
Background
Hypothyroidism results from a variety of abnormalities that cause insuf-
ficient secretion of thyroid hormones (see Table 1.18). The commonest
cause is autoimmune thyroid disease. Myxoedema is severe hypothyroid-
ism in which there is accumulation of hydrophilic mucopolysaccharides in
the ground substance of the dermis and other tissues, leading to thickening
of the facial features and a doughy induration of the skin. See Box 1.11 for
pitfalls with the thyroid function tests in non-thyroidal illness.
Epidemiology
• High TSH (>5.0) in 7.5% of ♀ and 2.5% of ♂ >65 years, 1.7% overt
hypothyroidism, 13.7% subclinical hypothyroidism (Whickham Survey, UK).
• Incidence higher in whites than Hispanics or African-American
populations.
• Incidence higher in areas of high iodine intake.
• An elevated TSH is associated with higher serum lipid concentrations,
which may be an additional reason to initiate therapy.
Clinical picture
Adult
• Insidious, non-specific onset.
• Fatigue, lethargy, constipation, cold intolerance, muscle stiffness, cramps,
carpal tunnel syndrome, menorrhagia, later oligo- or amenorrhoea.
• Slowing of intellectual and motor activities.
• d appetite and weight gain.
• Dry skin; hair loss.
• Deep hoarse voice, d visual acuity.
• Obstructive sleep apnoea.
Myxoedema
• Dull expressionless face, sparse hair, periorbital puffiness, macroglossia.
• Pale, cool skin that feels rough and doughy.
• Enlarged heart (dilation and pericardial effusion).
• Megacolon/intestinal obstruction.
• Cerebellar ataxia.
• Prolonged relaxation phase of deep tendon reflexes.
• Peripheral neuropathy.
• Encephalopathy.
• Hyperlipidaemia.
• Hypercarotenaemia (also caused by hyperlipidaemia, diabetes mellitus,
anorexia, and porphyria).
• Psychiatric symptoms, e.g. depression, psychosis.
• Marked respiratory depression with i arterial PCO2.
• Hyponatraemia from impaired water excretion and disordered
regulation of vasopressin secretion.
Myxoedema coma
• Predisposed to by cold exposure, trauma, infection, administration of
central nervous system depressants.
HYPOTHYROIDISM 75
Subclinical hypothyroidism
• This term is used to denote raised TSH levels in the presence of
normal concentrations of free thyroid hormones.
• Treatment is indicated if the biochemistry is sustained in patients
with a past history of radioiodine treatment for thyrotoxicosis or
+ve thyroid antibodies as, in these situations, progression to overt
hypothyroidism is almost inevitable (at least 5% per year of those with
+ve antithyroid peroxidase antibodies).
• Two samples should be taken 2–3 months apart to distinguish from
non-thyroidal illness.
• The reference range for serum TSH rises with age.
• There is controversy over the advantages of T4 treatment in patients
with –ve thyroid antibodies and no previous radioiodine treatment.
• If treatment is not given, follow-up with annual thyroid function tests is
important.
• There is no generally accepted consensus of when patients should
receive treatment. Some authorities suggest treatment when
serum TSH is >10mU/L because of i rate of progression to overt
hypothyroidism. Below that, if the TSH is raised, treatment is geared
to the individual patient.
• Increased incidence of cardiovascular risk, probably greater if
<65 years old.
• There is some evidence that women with autoimmune thyroiditis are
more likely to have an increased risk of recurrent miscarriage, although
it is not known whether the risk is related to thyroid autoimmunity or
to subtle thyroid failure. In this situation, there may be an advantage to
giving thyroxine if TSH is between 2.5 and 5mU/L.
• Acceleration of thyroid hormone metabolism during pregnancy can
lead to hypothyroidism in women with a limited thyroid hormone
reserve. If untreated, mild hypothyroidism can lead to subtle
impairment of subsequent childhood neuropsychological development.
Early correction of maternal hypothyroxinaemia is recommended,
aiming to maintain serum TSH in the lower half of the reference range
prior to conception, if possible.
Management
• If serum TSH is >10mU/L, then levothyroxine is indicated.
• If serum TSH is mildly increased between 4–10mU/L and the patient
is TPO antibody +ve, an annual check of serum TSH is recommended,
with commencement of levothyroxine once serum TSH >10mU/L.
• If the patient is thyroid antibody –ve, then ensuring a check of serum
TSH every 3–5 years is all that is required.
• If the patient with a serum TSH 4–10mU/L has symptoms consistent
with hypothyroidism and the TSH elevation persists, then a
3–6 months’ therapeutic trial of levothyroxine appears justified. If
the patient feels improved by therapy, it is reasonable to continue
treatment.
SUBCLINICAL HYPOTHYROIDISM 79
• If the patient with serum TSH 4–10U/L does not have symptoms and
the serum TSH level appears stable with +ve TPO antibodies, the
annual risk of progression to overt hypothyroidism is 4% per year,
and so an annual serum TSH surveillance strategy is warranted. If
the TPO antibodies are –ve, then 3-yearly serum TSH surveillance is
recommended, with a risk of progression to overt hypothyroidism of
3% per year.
• The exception to the above is in neonates and children, pregnancy, or
in someone trying to conceive when a mildly increased serum TSH
should always be treated, as it is associated with adverse outcomes for
both mother and fetus.
• Recent data suggest that levothyroxine treatment for those with serum
TSH 5–10U/L may also now be justified in subjects <65 years and in
those older subjects with documented evidence of heart failure on
echocardiography.
80 CHAPTER 1 Thyroid
Treatment of hypothyroidism
Note that patients in the UK with hypothyroidism are entitled to a medical
exemption certificate for prescription charges.
• Thyroid hormone replacement with synthetic levothyroxine remains
the treatment of choice in primary hypothyroidism. See Table 1.19 for
excipient content in case of intolerance.
• Levothyroxine with a long half-life enables the patient to take two
doses at once if a dose is omitted.
• Normal metabolic state should be restored gradually, as a rapid
increase in metabolic rate may precipitate cardiac arrhythmias.
• The average replacement dose is 1.6–1.8 micrograms/kg/day.
• To avoid iatrogenic subclinical disease, fasting ingestion of
levothyroxine is best either in the morning or the evening.
• In the younger patients, start levothyroxine at 50–100 micrograms. In
the elderly with a history of ischaemic heart disease, an initial dose of
levothyroxine 25–50 micrograms can be i by 25 micrograms increments
at 4-week intervals until normal metabolic state and TSH is attained.
• Optimum dose is determined by clinical criteria, the objective of
treatment being to restore serum TSH to the normal range.
• TSH should be checked only 6 weeks after any dose change. Once
stabilized, TSH should be checked on an annual basis. Patients should
stay on the same brand of levothyroxine as far as is possible—
otherwise, TSH should be checked at 2 months.
• Muscle problems may take up to 6 months to fully recover.
• It is estimated that 5% of hypothyroid patients remain symptomatic
in spite of levothyroxine replacement and normal serum TSH
concentrations. Possible causes include awareness of a chronic
disease, presence of associated autoimmune diseases, and thyroid
autoimmunity per se (independent of thyroid function).
• In those patients who remain symptomatic and in whom serum TSH
remains towards the upper end of the reference range, it is usual
practice for the dose of levothyroxine to be increased slightly to target
a serum TSH at the lower end of the reference range (≤2.5mU/L).
Whether or not such small changes in serum TSH within the reference
range are associated with symptoms is uncertain.
• Patients with iatrogenic hyperthyroidism (serum TSH <0.03U/L) have a
significantly increased risk of arrhythmia and fractures.
• In most trials, combination levothyroxine-T3 therapy does not appear
to be superior to levothyroxine monotherapy for the management of
hypothyroid symptoms. In three trials, patients preferred combined
therapy to levothyroxine monotherapy; however, in one of those
studies, patients were given doses of thyroid hormone which resulted
in mild hyperthyroidism. In general, clinical trials of combination
levothyroxine-T3 therapy have not successfully replicated physiological
T4-T3 production. Forty micrograms of T3 is approximately equivalent
to 0.125mg of T4.
• One study has suggested that a small group of patients with a
polymorphism in D2 may benefit from levothyroxine-T3 therapy.
Table 1.19 Excipient content of generic levothyroxine tablets
Manufacturer Goldshield Norton/IVAX Cox/Alpharma APS/Berk CP Pharmaceuticals Hillcross
(manufactured (50 + 100 (50 + 100 (only make 25 25 micrograms 50/
by Goldshield) micrograms micrograms micrograms) mic
Excipient only)* only)*
Thyroxine
Lactose
Sucrose
Dextrin
Maize starch
Magnesium stearate
Pre-gelatinized maize
starch
Stearic acid
Water (50 + 100
micrograms)
Sodium citrate
Powdered acacia
* 25 microgram tablets made by Goldshield.
Hillcross: 25 micrograms are manufactured by CP Pharmaceuticals; 50 and 100 micrograms are manufactured by Alpharma.
Norton/IVAX: 25, 50, and 100 micrograms are manufactured by Goldshield.
Data from Drug Information department of each manufacturer, August 2004.
82 CHAPTER 1 Thyroid
Question compliance
Confirm
Consider thyroxine absorption test +/– diagnosis and
supervised weekly dose administration treat
(single dose = 7 × 1.6 mcg/kg) with TSH
monitoring monthly.
Congenital hypothyroidism
Incidence—about 1 in 3,500–4,000 neonates. All neonates should be
screened. There is an inverse relationship between age at diagnosis and
intelligence quotient (IQ) in later life. In iodine-replete areas, 85% of the
cases are due to sporadic developmental defects of the thyroid gland (thy-
roid dysgenesis), such as the arrested migration of the embryonic thyroid
(ectopic thyroid) or a complete absence of thyroid tissue (athyreosis).
The remaining 15% have thyroid dyshormonogenesis defects transmitted
by an autosomal recessive mode of inheritance. See Box 1.13 for clinical
features.
Thyroid hormone dysgenesis
• Caused by inborn errors of thyroid metabolism. The disorders may be
autosomal recessive, indicating single protein defects.
• Can be caused by inactivation of the TSH receptor, abnormalities
of the thyroid transcription factors TTF1, TTF2, and PAX8, or due
to defects in iodide transport, organification (peroxidase), coupling,
deiodinase, or thyroglobulin synthesis.
• In a large proportion of patients with congenital hypothyroidism, the
molecular background is unknown.
Pendred’s syndrome
Characterized by overt or subclinical hypothyroidism, goitre, and
moderate-to-severe sensorineural hearing impairment. The prevalence
varies between 1 in 15,000 and 1 in 100,000. There is a partial iodide
organification defect detected by i perchlorate discharge. Thyroid hor-
mone synthesis is only mildly impaired and so may not be detected by
neonatal thyroid screening.
Laboratory tests
• Neonatal screening by measurement of serum TSH.
• Imaging procedure: ultrasonography or 123I scintigraphy.
• Measurement of serum thyroglobulin and low molecular weight
iodopeptides in urine to discriminate between the various types of
defects.
• Measurement of neonatal and maternal autoantibodies as an indication
of possible transient hypothyroidism.
Treatment
Irrespective of the cause of congenital hypothyroidism, early treatment
is essential to prevent cerebral damage. Sufficient T4 should be given to
maintain the TSH in the normal range.
Further reading
Biondi B, Cooper DC (2008). The clinical significance of subclinical thyroid dysfunction. Endocr
Rev 29, 76–131.
Escobar-Morales HF, Botella-Carretero JI, Escobar del Rey F, et al. (2005). Treatment of hypo-
thyroidism with combinations of levothyroxine plus liothyronine. J Clin Endocrinol Metab 90,
4949–54.
Flynn RW, Bonellie SR, Jung RT, et al. (2010). Serum thyroid-stimulating hormone concentration
and morbidity from cardiovascular disease and fractures in patients on long-term thyroxine
therapy. J Clin Endocrinol Metab 95, 186–93.
Garber JR, Cobin RH, Gharib H, et al. (2012). Clinical practice guidelines for hypothyroidism in
adults. Thyroid 22, 1–32.
Grozinsky-Glasberg S, Fraser A, Nahshoni E, et al. (2006). L. Thyroxine-triiodothyronine com-
bination therapy versus thyroxine monotherapy for clinical hypothyroidism: meta-analysis of
randomized controlled trials. J Clin Endocrinol Metab 91, 2592–9.
Grüters A, Krude H (2007). Update on the management of congenital hypothyroidism. Horm Res
68(Suppl 5), 107–111.
Hennemann G, Docter R, Visser TJ, et al. (2004). Thyroxine plus low-dose, slow-release triiodothy-
ronine replacement in hypothyroidism: proof of principle. Thyroid 14, 271–5.
Nygaard B, Jensen EW, Kvetny J, et al. (2009). Effect of combination therapy with thyroxine
(T4) and 3,5,3’-triiodothyronine versus T4 monotherapy in patients with hypothyroidism, a
double-blind, randomised cross-over study. Eur J Endocrinol 161, 895–902.
Okosieme OE, Belludi G, Spittle K, et al. (2011). Adequacy of thyroid hormone replacement in a
general population. QJM 104, 395–401.
Roberts CG, Ladenson PW (2004). Hypothyroidism. Lancet 363, 793–803.
Rodondi N, den Elzen WP, Bauer DC, et al. (2010). Subclinical hypothyroidism and the risk of
coronary heart disease and mortality. JAMA 304, 1365–74.
Royal College of Physicians (2011). The diagnosis and management of primary hypothyroidism.
Royal College of Physicians, London.
Taylor PN, Panicker V, Sayers A, et al. (2011). A meta-analysis of the associations between com-
mon variation in the PDE8B gene and thyroid hormone parameters, including assessment of
longitudinal stability of associations over time and effect of thyroid hormone replacement. Eur
J Endocrinol 164, 773–80.
Vaidya B, Pearce SH (2008). Management of hypothyroidism. BMJ 337, a801.
Vanderpump MP (2010). How should we manage patients with mildly increased serum thyrotro-
phin concentrations? Clin Endocrinol (Oxf) 72, 436–40.
86 CHAPTER 1 Thyroid
Further reading
Bogazzi F, Bartalena L, Dell’Unto E, et al. (2007). Proportion of type 1 and type 2 amiodarone-induced
thyrotoxicosis has changed over a 27-year period in Italy. Clin Endocrinol (Oxf) 67, 533–7.
Bogazzi F, Bartalena L, Martino E (2010). Approach to the patient with amiodarone-induced thyro-
toxicosis. J Clin Endocrinol Metab 95, 2529–35.
Bogazzi F, Tomisti L, Rossi G, et al. (2009). Glucocorticoids are preferable to thionamides as
first-line treatment for amiodarone-induced thyrotoxicosis due to destructive thyroidi-
tis: A matched retrospective cohort study. J Clin Endocrinol Metab 94, 3757–62.
Franklyn JA, Gammage MD (2007). Treatment of amiodarone-associated thyrotoxicosis. Nat Clin
Pract Endocrinol Metab 3, 662–6.
Han T, Williams GR, Vanderpump MP (2009). Benzofuran derivatives and the thyroid. Clin
Endocrinol (Oxf) 70, 2–13.
Loy M, Perra E, Mellis A, et al. (2007). Color-flow doppler sonography in the differential diagnosis
and management of amiodarone-induced thyrotoxicosis. Acta Radiol 48, 628–34.
Tanda ML, Piantanida E, Lai A, et al. (2008). Diagnosis and management of amiodarone-induced
thyrotoxicosis: similarities and differences between North American and European thyroidolo-
gists. Clin Endocrinol (Oxf) 69, 812–18.
EPIDEMIOLOGY OF THYROID CANCER 91
Table 1.26 TNM staging system for papillary and follicular thyroid
carcinoma
Stage Age <45 years Age >45 years
I Any T, any N, M0 T1, N0, M0
II Any T, any N, M1 T2, N0, M0
III T3, N0, M0 or any T1–3, N1a, M0
IVA T1–3, N1b, M0 or T4a, any N, M0 N, M0
IVB T4b, any N, M0
IVC Any T, any N, M1
Primary tumour (T): T1, tumour ≤ 2cm limited to the thyroid; T2, tumour > 2 to ≤ 4cm
limited to the thyroid; T3, tumour > 4cm limited to the thyroid or any tumour with minimal
extrathyroidal extension (e.g. extension to sternothyroid muscle or perithyroidal soft tissues);
T4a, tumour of any size with extension beyond the thyroid capsule and invading any of the
following: subcutaneous soft tissues, larynx, trachea, oesophagus, recurrent laryngeal nerve;
T4b, tumour invading prevertebral fascia, mediastinal vessels, or encases carotid artery.
Lymph nodes (N): To classify as N0 or N1, at least six lymph nodes should be examined at
histology. Otherwise, the tumour is classified as Nx. N0, no regional lymph node metastasis;
N1a, metastases in pretracheal and paratracheal, including prelaryngeal and Delphian lymph
nodes; N1b, metastases in other unilateral, bilateral, or contralateral cervical or upper
mediastinal lymph nodes.
Distant metastases (M): M0, no distant metastasis; M1, distant metastasis.
Reproduced from Wass, J, Oxford Textbook of Endocrinology and Diabetes, 2011, with
permission from OUP.
prior to the scan, allowing TSH to rise. A low-iodine diet for 2 weeks
increases the effective specific activity of the administered iodine.
Exogenous TSH is an alternative to T3 cessation.
• The patient should be isolated until residual dose meter readings
indicate <30MBq.
• Chronic suppression of serum TSH levels to <0.10mU/L is standard
practice in patients with differentiated thyroid carcinoma. Inhibition of
TSH secretion reduces recurrence rate, as TSH stimulates growth of
the majority of thyroid cancer cells.
Patients are followed up with thyroglobulin levels. After effective treat-
ment, thyroglobulin levels are undetectable. A trend of i thyroglobulin
values should be investigated with a radioiodine uptake scan. Liothyronine
(T3) is substituted for T4 4–6 weeks before the scan and omitted for 10 days
immediately beforehand. As above exogenous TSH can also be used.
Thyroglobulin
• A very sensitive marker of recurrence of thyroid cancer.
• Secreted by the thyroid tissue.
• After total thyroidectomy and radioactive iodine ablation, the levels of
thyroglobulin should be <0.27 micrograms/L.
• Measurement of thyroglobulin levels could be made difficult in the
presence of antithyroglobulin antibodies, which should be checked.
• Coming off thyroid hormones or giving recombinant TSH increases
the sensitivity of thyroglobulin to detect recurrence, but this may not
affect survival rates.
Recurrent disease/distant metastases
• In the case of recurrence, treatment employs all methods used in p
and adjuvant therapy.
• Surgery for local metastases.
• Radioactive iodine for those tumours with uptake.
• External radiotherapy is indicated in non-resectable tumours that do
not take up 131I.
• Bony and pulmonary metastases (usually osteolytic) may be treated
with 131I.
• Unfortunately, only 50% of metastases concentrate 131I.
• External beam radiation is given to patients who have gross residual
disease after attempted surgery and radioiodine. Radiation therapy
may be of value in controlling local disease. If thyroidectomy is not
possible, it is given alone for palliation.
• Due to the low efficacy of traditional cytotoxic chemotherapies,
consensus guidelines now recommend consideration of clinical trials
of novel agents when patients require therapy for progressive, locally
advanced, or metastatic differentiated thyroid cancer.
• Tyrosine kinase inhibitors (TKIs) (e.g. sorafenib, sunitinib, pazopanib,
gefitinib) have been of interest for the treatment of advanced
differentiated thyroid cancer, given the oncogenic roles of mutations
in the serine kinase BRAF and tyrosine kinases RET (in the mutated
fusion protein RET/PTC) and RAS and the contributory roles of tyrosine
98 CHAPTER 1 Thyroid
Tg undetectable Tg detectable
Positive
CT
Further imaging
PET
Fig. 1.3 Algorithm for follow-up of low-risk cases of papillary thyroid cancer.
Anaplastic (undifferentiated)
thyroid cancer
• Rare.
• Peak incidence: seventh decade; ♀:♂ = 1:1.5.
• Characterized by rapid growth of a firm/hard, fixed tumour.
• Often infiltrates local tissue, such as larynx and great vessels, and so
does not move on swallowing. Stridor and obstructive respiratory
symptoms are common.
• Aggressive, with poor long-term prognosis—7% 5-year survival rate
and a mean survival of 6 months from diagnosis.
• Optimal results occur, following total thyroidectomy. This is usually
not possible and external irradiation is used, sometimes in association
with chemotherapy.
LYMPHOMA 105
Lymphoma
• Uncommon.
• Almost always associated with autoimmune thyroid disease
(Hashimoto’s thyroiditis). Occurs more commonly in ♀ and in patients
aged >40 years.
• Characterized by rapid enlargement of the thyroid gland.
• May be limited to thyroid gland or part of a more extensive systemic
lymphoma (usually non-Hodgkin’s lymphoma).
• Treatment with radiotherapy alone or chemotherapy, if more
extensive, often produces good results.
Further reading
American Thyroid Association Guidelines Task Force (2009). Medullary thyroid cancer: manage-
ment guidelines of the American Thyroid Association. Thyroid 19, 565–612.
American Thyroid Association (ATA) Guidelines Taskforce on Thyroid Nodules and Differentiated
Thyroid Cancer (2009). Revised American Thyroid Association management guidelines for
patients with thyroid nodules and differentiated thyroid cancer. Thyroid 19, 1167–214.
Aschebrook-Kilfoy B, Ward MH, Sabra MM, et al. (2011). Thyroid cancer incidence patterns in the
United States by histologic type, 1992-2006. Thyroid 21, 125–34.
Bogsrud TV, Hay ID, Karantanis D, et al. (2011). Prognostic value of 18F-fluorodeoxyglucose-positron
emission tomography in patients with differentiated thyroid carcinoma and circulating antithy-
roglobulin autoantibodies. Nucl Med Commun 32, 245–51.
British Thyroid Association and Royal College of Physicians (2007). Guidelines for the management
of thyroid cancer (Perros P, ed). Report of the Thyroid Cancer Guidelines Update Group. Royal
College of Physicians, London. Available at: M http://www.british-thyroid-association.org/news/
Docs/Thyroid_cancer_guidelines_2007.pdf.
Hay ID (2007). Management of patients with low-risk papillary thyroid carcinoma. Endocr Pract
13, 521–33.
Hay I, Wass JAH (2008). Clinical Endocrine Oncology, 2nd edn, pp.109–71. Blackwell-Wiley,
Massachussetts.
Kloos RT, Eng C, Evans D, et al. (2009). Medullary thyroid cancer: management guidelines of the
American Thyroid Association. Thyroid 19, 565–612.
Nikiforov YE (2006). Radiation-induced thyroid cancer: what we have learned from Chernobyl.
Endocr Pathol 17, 307–17.
Nikiforov YE, Nikiforova MN (2011). Molecular genetics and diagnosis of thyroid cancer. Nat Rev
Endocrinol 7, 569–80.
Paschke R, Hegedüs L, Alexander E, et al. (2011). Thyroid nodule guidelines: agreement, disagree-
ment and need for future research. Nat Rev Endocrinol 7, 354–61.
Sherman SI (2011). Targeted therapies for thyroid tumors. Mod Pathol 24(Suppl 2), S44–52.
Smallridge RC (2012). Approach to the patient with anaplastic thyroid carcinoma. J Clin Endocrinol
Metab 97, 2566–72.
Anatomy and physiology of anterior pituitary gland 108
Imaging 112
Pituitary function—dynamic tests: insulin tolerance test
(ITT) 114
Glucagon test 116
ACTH stimulation test 117
Arginine test 118
Clomifene test 119
hCG test 120
TRH test 121
Hypopituitarism 122
Investigations of hypopituitarism 125
Treatment of hypopituitarism 126
Anterior pituitary hormone replacement 127
Glucocorticoids 128
Growth hormone (GH) replacement therapy in adults 130
Investigation of GH deficiency 132
Treatment of GH deficiency 134
Pituitary tumours 136
Molecular mechanisms of pituitary tumour pathogenesis 138
Prolactinomas 140
Investigations of prolactinomas 142
Hyperprolactinaemia and drugs 143
‘Idiopathic’ hyperprolactinaemia 143
Treatment of prolactinomas 144
Definition of acromegaly 148
Epidemiology of acromegaly 148
Causes of acromegaly 149
Associations of acromegaly 150
Clinical features of acromegaly 151
Investigations of acromegaly 152
Management of acromegaly 154
Mortality data of acromegaly 157
Chapter 2 107
Pituitary
Supraoptic recess
Suprasellar Hypothalamus
cistern
Optic
Sphenoid chiasm
bone Pituitary stalk
III Temporal
IV lobe
VI Internal
carotid
V2 VI
artery
Fig. 2.1 The pituitary gland. Reproduced with permission from Weatherall DJ,
Ledingham JGG, and Warrell DA (eds) (1996). Oxford Textbook of Medicine, 3rd
edn. Oxford University Press: Oxford.
*
Concentrations i with stress: NB venepuncture.
Table 2.1 Anterior pituitary gland physiology
Cell type (hormone) % pituitary +ve regulation –ve regulation Targets Effects
Somatotropes (growth 45–50 Growth hormone-releasing Insulin-like growth factor Liver, cartilage, Linear and somatic
hormone, GH) hormone (GHRH) (IGF-1) and somatostatin muscle, fat, skin Metabolism (lipids,
carbohydrates)
Lactotropes (prolactin, 15–25 (i in Thyrotropin-releasing Dopamine Breast Lactation
PRL) pregnancy) hormone (TRH) and
oestrogen
Gonadotropes 10–15 Gonadotrophin-releasing Oestrogen Gonads Sex steroid product
(luteinizing hormone hormone (GnRH),
Progesterone Folliculogenesis and
and follicle-stimulating oestrogen—late follicular
ovulation (♀)
hormone, LH/FSH) phase of menstrual cycle Testosterone
Spermatogenesis (♂
Inhibin (FSH only)
Thyrotropes 5–10 TRH T4, T3, somatostatin Thyroid Thyroid hormone
(thyroid-stimulating production
hormone, TSH)
Corticotropes 15–20 Corticotrophin-releasing Cortisol Adrenal gland Glucocorticoid and
(adrenocorticotrophin, hormone (CRH) production
ACTH)
Reproduced from Draznin and Epstein, Oxford American Handbook of Endocrinology and Diabetes (2011), with permission of OUP.
ANATOMY AND PHYSIOLOGY OF ANTERIOR PITUITARY GLAND 111
112 CHAPTER 2 Pituitary
Imaging
Background
• Magnetic resonance imaging (MRI) currently provides the optimal
imaging of the pituitary gland.
• Computed tomography (CT) scans may still be useful in demonstrating
calcification in tumours (e.g. craniopharyngiomas) and hyperostosis in
association with meningiomas or evidence of bone destruction.
• Plain skull radiography may show evidence of pituitary fossa
enlargement but has been superseded by MRI.
MRI appearances
(See Fig. 2.2.)
• T1-weighted images demonstrate cerebrospinal fluid (CSF) as dark
grey and brain as much whiter. This imaging is useful for demonstrating
anatomy clearly. The normal posterior pituitary gland appears
bright white (due to neurosecretory granules and phospholipids) on
T1-weighted images, in contrast to the anterior gland which is of the
same signal as white matter. The bony landmarks have low signal
intensity on MRI, and air in the sphenoid sinus below the fossa shows
no signal. Fat in the dorsum sellae may shine white. T2-weighted
images may sometimes be used to characterize haemosiderin and fluid
contents of a cyst.
• IV gadolinium compounds are used for contrast enhancement. Because
the pituitary and pituitary stalk have no blood–brain barrier, in contrast
to the rest of the brain, the normal pituitary gland enhances brightly
following gadolinium injection. Contrast enhancement is particularly
useful for the demonstration of cavernous sinus involvement and of
microadenomas.
• The normal pituitary gland has a flat or slightly concave upper surface.
In adolescence or pregnancy, the surface may become slightly convex.
Pituitary adenomas
On T1-weighted images, pituitary adenomas are of lower signal intensity
than the remainder of the normal gland. The size and extent of the pitui-
tary adenomas are noted in addition to the involvement of other struc-
tures, such as invasion of the cavernous sinus, erosion of the fossa, and
relations to the optic chiasm. Larger tumours may show low-intensity
areas compatible with necrosis or cystic change or higher intensity signal
due to haemorrhage. The presence of microadenomas may be difficult
to demonstrate. Contrast enhancement, asymmetry of the gland, or stalk
position can be helpful in such cases.
Neuroradiological classification
b see p. 184.
IMAGING 113
(A) (B)
R L
OC
OC
PS
(C) (D)
OC
PG
CA
Fig. 2.2 Normal and abnormal pituitary MRI images. (A) Normal coronal,
post-contrast T1 image showing optic chiasm (OC). (B) Normal sagittal image
showing pituitary stalk (PS) and OC. (C) Pituitary macroadenoma with evidence of
pituitary gland (PG) compression, left cavernous sinus invasion, and encroachment
of carotid artery (CA). (D) Rathke’s cleft cyst—bright T1 image, consistent with
mucinous, proteinaceous, or blood products. Reproduced from Draznin and
Epstein, Oxford American Handbook of Endocrinology and Diabetes (2011), with
permission of OUP.
Craniopharyngiomas
These appear as intra- and/or suprasellar masses with cystic and/or solid
components. A solid lesion appears as iso- or hypointense relative to the
brain on pre-contrast T1-weighted images, shows enhancement following
gadolinium administration, and is usually of mixed hypo- or hyperinten-
sity on T2-weighted sequences. A cystic element is usually hypointense on
T1- and hyperintense on T2-weighted sequences. Protein, cholesterol, and
methaemoglobin may cause high signal on T1-weighted images. Calcification
is present in 45–57%, better visualized by CT or plain skull X-ray.
Further reading
Naidich MJ, Russell EJ (1999). Current approaches to imaging of the sellar region and pituitary.
Endocrinol Metab Clin N Am 28, 45.
114 CHAPTER 2 Pituitary
Pituitary function—dynamic
tests: insulin tolerance test (ITT)
Indications
• Assessment of ACTH reserve.
• Assessment of GH reserve.
Physiology
IV insulin is used to induce hypoglycaemia (glucose <2.2mmol/L with signs
of glycopenia), which produces a standard stress causing ACTH and GH
secretion.
Contraindications
• 9 a.m. serum cortisol <100nmol/L.
• Untreated hypothyroidism.
• Abnormal ECG.
• Ischaemic heart disease.
• Seizures.
• Glycogen storage disease.
Note that patients should discontinue oral oestrogen replacement for 6
weeks before the test, as i CBG will make the cortisol results difficult
to interpret. Progesterone and transdermal oestrogen may be continued.
See Box 2.1 for procedure.
Response
• In the presence of inadequate hypoglycaemia (glucose >2.2mmol/L),
the test cannot be interpreted.
• A normal cortisol response (peak cortisol >450nmol/L) demonstrates
the ability to withstand stress (including major surgery) without
requiring glucocorticoid cover. Subnormal cortisol response requires
glucocorticoid replacement treatment (b see Glucocorticoids, p. 128).
• Severe GH deficiency in adults is diagnosed if peak GH <3 micrograms/L,
and, in the appropriate clinical situation (b see p. 130), GH replacement
therapy may be recommended. In children, the secretory capacity of GH
is higher, and a cut-off of 10 micrograms/L is used.
PITUITARY FUNCTION—DYNAMIC TESTS: ITT 115
Glucagon test
Indications
• Assessment of ACTH reserve.
• Assessment of GH reserve.
Physiology
Glucagon leads to release of insulin, which then leads to GH and ACTH
release. The response may be s to the drop in glucose seen after the
initial rise following glucagon injection or may relate to the nausea induced
by glucagon.
Contraindications
• Phaeochromocytoma or insulinoma, glycogen storage disease, and
severe hypocortisolaemia.
• Often unreliable in patients with diabetes mellitus.
• Note that patients should discontinue oral oestrogen replacement
for 6 weeks before the test, as i CBG will make the cortisol results
difficult to interpret.
Response
• The normal response is a rise in glucose to a maximum at 90min. The
cut-offs for cortisol and GH are those of the ITT.
• This is a less reliable test than the ITT, as 20% of normal individuals
may fail to respond.
See Box 2.2 for procedure.
Arginine test1
Indication
Second-line test for assessment of GH reserve.
Physiology
Arginine leads to GH release.
Contraindications
None.
See Box 2.4 for procedure.
Response
A normal response is a rise in GH to >15–20mU/L.
Reference
1. Corneli G, Di Somma C, Baldelli R, et al. (2005). The cut-off limits of the GH response to
GH-releasing hormone–arginine test related to body mass index. European Journal of
Endocrinology, 153, 257–64.
CLOMIFENE TEST 119
Clomifene test
Indications
Assessment of gonadotrophin deficiency (e.g. Kallmann’s syndrome).
Physiology
Clomiphene has mixed oestrogen and antioestrogenic effects. The basis of
the test is competitive inhibition of oestrogen binding at the hypothalamus
and pituitary gland, leading to i LH and FSH after 3 days.
Contraindications
• Avoid in those with liver disease.
• May transiently worsen depression.
See Box 2.5 for procedure.
Response
• The normal response is a doubling of gonadotrophins by day 10,
usually rising beyond the normal range.
• In hypothalamic or pituitary disease, no gonadotrophin rise is seen.
Prepubertal patients may show a fall in gonadotrophins.
• Ovulation is presumed if day 21 progesterone is >30nmol/L.
120 CHAPTER 2 Pituitary
hCG test
Indications
To examine Leydig cell function, b see Clinical assessment, p. 402.
TRH test
Indications
• Differentiation of pituitary TSH and hypothalamic TRH deficiency.
• Differentiation of TSH-secreting tumour from thyroid hormone
resistance (b see Table 1.15, p. 51).
See Box 2.6 for procedure.
Response
• Normal response is a rise in TSH by >2mU/L to >3.4mU/L, with a
maximum at 20min and lower values at 60min.
• A delayed peak (60min rather than 20min) is typically found in
hypothalamic disease.
Hypopituitarism
Definition
Hypopituitarism refers to either partial or complete deficiency of anterior
and/or posterior pituitary hormones and may be due to p pituitary dis-
ease or to hypothalamic pathology which interferes with the hypothalamic
control of the pituitary.
Causes
• Pituitary tumours.
• Parapituitary tumours—craniopharyngiomas, meningiomas, secondary
deposits (e.g. breast, lung), chordomas, gliomas.
• Radiotherapy—pituitary, cranial, nasopharyngeal.
• Pituitary infarction (apoplexy), Sheehan’s syndrome.
• Infiltration of the pituitary gland—sarcoidosis, lymphocytic
hypophysitis, haemochromatosis, Langerhans cell histiocytosis,
Erdheim–Chester disease, Wegener’s (ANCA-positive, 1% pituitary
involvement, diabetes insipidus commonest).
• Empty sella.
• Infection—tuberculosis, pituitary abscess.
• Trauma (including traumatic brain injury).
• Subarachnoid haemorrhage.
• Isolated hypothalamic-releasing hormone deficiency, e.g. Kallmann’s
syndrome due to GnRH deficiency.
• Genetic causes.
• Mutations of genes encoding transcription factors, including HESX1
(homeobox gene expressed in embryonic stem cells 1), LHX3
(Lim-domain homeobox gene 3), LHX4 (Lim-domain homeobox
gene 4), PROP-1 (Prophet of Pit1), POU1F1 (Pou domain, class 1,
transcription factor 1).
• Russell viper envenomation.
Features
(See Table 2.2.)
• The clinical features depend on the type and degree of the hormonal
deficits, and the rate of its development, in addition to whether there
is intercurrent illness. In the majority of cases, the development of
hypopituitarism follows a characteristic order, with secretion of GH,
then gonadotrophins being affected first, followed by TSH and ACTH
secretion at a later stage. PRL deficiency is rare, except in Sheehan’s
syndrome associated with failure of lactation. ADH deficiency is
virtually unheard of with pituitary adenomas but may be seen rarely
with infiltrative disorders and trauma.
• The majority of the clinical features are similar to those occurring
when there is target gland insufficiency. There are important
differences, e.g. lack of pigmentation and normokalaemia in ACTH
deficiency in contrast to i pigmentation and hyperkalaemia (due to
aldosterone deficiency) in Addison’s disease.
• NB Houssay phenomenon. Amelioration of diabetes mellitus in patients
with hypopituitarism due to reduction in counter-regulatory hormones.
HYPOPITUITARISM 123
Apoplexy
Apoplexy refers to infarction of the pituitary gland due to either haemor-
rhage or ischaemia. It occurs most commonly in patients with pituitary
adenomas, usually macroadenomas, but other predisposing conditions
include post-partum (Sheehan’s syndrome), radiation therapy, diabetes
mellitus, anticoagulant treatment, disseminated intravascular coagulopa-
thy, and reduction in intracranial pressure. It is a medical emergency, and
rapid hydrocortisone replacement can be lifesaving.
It may present with a syndrome which is difficult to differentiate from
any other intracranial haemorrhage, with sudden onset headache, vomit-
ing, meningism, visual disturbance, and cranial nerve palsy. The diagnosis is
based on the clinical features and pituitary imaging which shows high signal
on T1- and T2-weighted images (b see p. 112). It should be managed by
a pituitary multidisciplinary team. It has been suggested that early surgery
(within 8 days) provides the optimal chance for neurological recovery.
However, some patients may be managed conservatively if the patient has
no significant visual or other neurological manifestations.
After apoplexy, pituitary tumour regrowth may occur (11% at 7 years),
so follow-up surveillance is necessary.
Empty sella syndrome
An enlarged pituitary fossa, which may be p (due to arachnoid herniation
through a congenital diaphragmatic defect) or s to surgery, radiotherapy,
or pituitary infarction. The majority of patients have normal pituitary func-
tion. Hypopituitarism (and/or hyperprolactinaemia) is found in <10%.
Traumatic brain injury (TBI)
• Associated with subarachnoid haemorrhage and skull base fractures.
• May be seen with moderate-to-severe head trauma.
• Most common deficiencies—GH and gonadotrophins (10–15%).
• Best assessed 6–12 months after trauma.
Sheehan’s syndrome
Haemorrhagic infarction of the enlarged post-partum pituitary gland caus-
ing hypopituitarism, following severe hypotension usually due to blood
loss, e.g. post-partum haemorrhage. It can be fatal, and survivors require
life replacement therapy. Improvement in obstetric care has made this a
rare occurrence in the developed world.
INVESTIGATIONS OF HYPOPITUITARISM 125
Investigations of hypopituitarism
The aims of investigation of hypopituitarism are to biochemically assess
the extent of pituitary hormone deficiency and also to elucidate the cause.
Basal hormone levels
Basal concentrations of the anterior pituitary hormone, as well as the tar-
get organ hormone, should be measured, as the pituitary hormones may
remain within the normal range despite low levels of target hormone.
Measurement of the pituitary hormone alone does not demonstrate that
the level is inappropriately low, and the diagnosis may be missed.
• LH and FSH, and testosterone (9 a.m.) or oestradiol.
• TSH and thyroxine.
• 9 a.m. cortisol.
• PRL.
• IGF-1 (NB May be normal in up to half of GHD, depending on age).
See Box 2.7 for dynamic tests.
Posterior pituitary function
• It is important to assess and replace corticotroph function before
assessing posterior pituitary hormone production because ACTH
deficiency leads to reduced GFR and the inability to excrete a water
load, which may, therefore, mask diabetes insipidus (DI).
• Plasma and urine osmolality are often adequate as baseline measures.
However, in patients suspected to have DI, a formal fluid deprivation
test should usually be performed (b see Box 2.31, p. 213).
Investigation of the cause
• Pituitary imaging—MRI ± contrast.
• Investigation of hormonal hypersecretion if a pituitary tumour is
demonstrated.
• Investigation of infiltrative disorders, (b see Parasellar inflammatory
conditions, pp. 192–3), e.g. serum and CSF ACE, ferritin, hCG, aFP.
• Occasionally, biopsy of a lesion found on imaging is required.
Treatment of hypopituitarism
Treatment involves adequate and appropriate hormone replacement
(b see p. 127, p. 128, and p. 130) and management of the underlying cause.
Isolated defects of pituitary hormone secretion
Rarely, patients have isolated insufficiency of only one anterior pituitary
hormone. The aetiology of these disorders is largely unknown, although
loss of hypothalamic control may play a role; an autoimmune pathology
has been suggested in some and genetic mutations in others. Examples
include:
• GnRH deficiency (Kallman’s syndrome—congenital GnRH deficiency
± anosmia).
• Isolated ACTH deficiency.
• Pit1 gene mutation (leads to isolated GH, PRL, and TSH deficiency).
• Prop1 gene mutation (leads to isolated GH, PRL, TSH, and
gonadotrophin deficiency).
Further reading
De Marinis L, et al. (2005). Primary empty sella. J Clin Endocrinol Metab 90, 5471–7.
Glynn N, Agha A (2013). Which patient requires neuroendocrine assessment following traumatic
brain injury, when and how. Clin Endocrinol 78, 17–20.
Pal A, et al. (2011). Pituitary apoplexy in non-functioning pituitary adenomas: long term follow up
is important because of significant numbers of tumour recurrences. Clin Endocrinol 75, 501.
Rajasekaran S (2011). UK guidelines for the management of pituitary apoplexy.Clini Endocrinol 74, 9.
Toogood AA, Stewart PM (2008). Hypopituitarism: clinical features, diagnosis, and management.
Endocrinol Metab Clin North Am 37, 235–61.
ANTERIOR PITUITARY HORMONE REPLACEMENT 127
Glucocorticoids
Replacement therapy
Patients with ACTH deficiency usually need glucocorticoid replacement
only and do not require mineralocorticoids, in contrast to patients with
Addison’s disease.
The normal production rate of cortisol is 9.9 ± 2.7mg/day. The gluco-
corticoid most commonly used for replacement therapy is hydrocortisone.
It is rapidly absorbed, with a short half-life (90–120min). Prednisolone and
dexamethasone can occasionally be used for glucocorticoid replacement.
The longer half-lives of these two drugs make them useful where sustained
ACTH suppression is required in, for example, congenital adrenal hyper-
plasia (CAH). Dexamethasone is useful when monitoring endogenous
production, as it is not detected in most cortisol assays. Cortisol acetate
was previously used for glucocorticoid replacement therapy but requires
hepatic conversion to active cortisol.
Monitoring of replacement
This is important to avoid over-replacement which is associated with
i BP, elevated glucose and insulin, and reduced bone mineral density
(BMD). Under-replacement leads to the non-specific symptoms, as seen
in Addison’s disease (b see p. 266). A clinical assessment is important,
but biochemical monitoring, using plasma and urine cortisol (UFC) meas-
urements, is used by many endocrinologists. The aim is to keep the UFC
within the reference range. Many centres use plasma cortisol measure-
ments on a hydrocortisone day curve (see Box 2.8). The aim is to keep the
plasma cortisol between 150 and 300nmol/L, avoiding nadirs of <50nmol/L
predose. Conventional replacement (20mg hydrocortisone/24h) may
overtreat patients with partial ACTH deficiency.
Safety
Patients should be encouraged to wear a MedicAlert, indicating that they
are cortisol-deficient, to carry a steroid card, and to keep a vial of paren-
teral hydrocortisone at home to be administered in emergency situations.
Equivalent oral glucocorticoid doses
1mg hydrocortisone is equivalent to:
• 1.5mg cortisol acetate.
• 0.2mg prednisolone.
• 0.0375mg dexamethasone.
• 4mg prednisolone ≡ 20mg hydrocortisone ≡ 0.075mg dexamethasone.
GLUCOCORTICOIDS 129
Investigation of GH deficiency
Dynamic tests of GH secretion
• ITT is most widely used test. Peak GH <10mU/L (3 micrograms/L) is
diagnostic of severe GHD (see b p. 114).
• Alternative tests, if the ITT is contraindicated, include a combination
of GHRH (1 microgram/kg) and arginine (0.5g/kg) IV over 30min
(see b p. 118).
• A second confirmatory dynamic biochemical test is recommended,
particularly in patients who have suspected isolated GH deficiency.
A single GH dynamic test is sufficient to diagnose GHD in patients with
two or three pituitary hormonal defects.
IGF-1
IGF-1 concentrations may remain within the age-matched reference range
despite severe GHD in up to 50% of patients and, therefore, do not
exclude the diagnosis, and reduced IGF-1 is seen in a number of conditions.
For causes of lowered IGF-1 levels, see Box 2.9.
Abnormalities in adult GH deficiency
• Stimulated GH <10mU/L (3 micrograms/L).
• Low or low-normal IGF-1 (IGF-1 may be normal in up to 50%,
depending on age).
• d BMD.
• i insulin resistance.
• Dyslipidaemia (i LDL).
• Impaired cardiac function.
Clinical features of GH deficiency
• Impaired well-being.
• Reduced energy and vitality (depressed mood, i social isolation,
i anxiety).
• Reduced muscle mass and impaired exercise capacity.
• i central adiposity (i waist/hip ratio) and i total body fat.
• d sweating and impaired thermogenesis.
• i cardiovascular risk.
• i fracture risk (osteoporosis).
Treatment of GH deficiency
All patients with GHD should be considered for GH replacement therapy.
In particular, patients with impaired QoL, reduced mineral density, an
adverse cardiovascular risk profile, and reduced exercise capacity should
be considered for treatment.
Dose
See Box 2.10.
• Unlike paediatric practice, where GH doses are determined by body
weight and surface area, most adult endocrinologists use dose titration,
using serial IGF-1 measurements, to increase the dose of GH until the
IGF-1 level approaches the middle to upper end of the age-matched
IGF-1 reference range. This reduces the likelihood of side effects,
mainly related to fluid retention, which were frequently observed in
the early studies of GH replacement in adults when doses equivalent
to those used in paediatric practice were used.
• The normal production of GH is 200–500 micrograms/day in an adult.
Current recommendations are a starting dose of 150–300 micrograms/
day. The maintenance dose is usually 200–600 micrograms/day. The
dose in ♀ is often higher than for age-matched ♂ (particularly if the
female is on oral oestrogens).
Monitoring of treatment
• A clinical examination, looking for reduction in overall body weight
(a good response is loss of 3–5kg in 12 months) and reduced waist/
hip ratio. BP may fall in hypertensive patients because of reduction in
peripheral systemic vascular resistance.
• IGF-1 is monitored to avoid over-replacement, aiming to keep values
within the age-matched reference range. During dose titration, IGF-1
should be measured every 1–2 months. Once a stable dose is reached,
IGF-1 should be checked at least once a year.
• The adverse effects experienced with GH replacement usually resolve
with dose reduction and tend to be less frequent with the lower
starting doses used in current practice.
• GH treatment may be associated with impairment of insulin sensitivity,
and therefore markers of glycaemia should be monitored.
• Lipids should be monitored annually. BMD should be monitored every
2 years, particularly in those with d BMD.
• It may be helpful to monitor QoL using a questionnaire, such as the
AGHDA (adult GHD assessment) questionnaire.
Further reading
Carroll PV, Christ ER, Bengtsson BA, et al. (1998). GH deficiency in adulthood and the effects of
GH replacement: a review. J Clin Endocrinol Metab 83, 382–95.
Molitch ME, Clemmons DR, Malozowski S, Merriam GR, Vance ML (2011). Endocrine Society.
Evaluation and treatment of adult growth hormone deficiency: an Endocrine Society clinical
practice guideline. J Clin Endocrinol Metab 96, 1587–609.
136 CHAPTER 2 Pituitary
Pituitary tumours
Epidemiology
• Pituitary adenomas are the most common pituitary disease in adults
and constitute 10–15% of primary brain tumours.
• Approximately 10% of individuals harbour incidental tumours, most
commonly microadenomas.
• The incidence of clinically apparent pituitary disease is 1 in 10,000.
• Pituitary carcinoma is very rare (<0.1% of all tumours) and is most
commonly ACTH- or prolactin-secreting.
See Table 2.4.
Classification
Size
• Microadenoma <1cm.
• Macroadenoma >1cm.
Functional status (clinical or biochemical)
• Prolactinoma 35–40%.
• Non-functioning 30–35%.
• Growth hormone (acromegaly) 10–15%.
• ACTH adenoma (Cushing’s disease) 5–10%.
• TSH adenoma <5%.
Pathogenesis
The mechanism of pituitary tumourigenesis remains largely unclear.
Pituitary adenomas are monoclonal, supporting the theory that there are
intrinsic molecular events leading to pituitary tumourigenesis. However,
the mutations (e.g. p53) found in other tumour types are only rarely
found. A role for hormonal factors and, in particular, the hypothalamic
hormones in tumour progression is also a suggested hypothesis.
Mortality
Pituitary disease is associated with an increased mortality, predominantly
due to vascular disease. This may be due to oversecretion of GH or
ACTH, hormone deficiencies or excessive replacement (e.g. of hydrocor-
tisone). Radiotherapy of the pituitary is also associated with an increased
mortality (especially cerebrovascular). Craniopharyngioma patients have a
particularly increased mortality.
Reference
1. Fernandez, et al. (2010). Prevalence of pituitary adenomas: a community-based, cross-sectional
study in Banbury (Oxfordshire, UK). Clin Endocrinol (Oxf) 72, 377.
138 CHAPTER 2 Pituitary
Prolactinomas
Epidemiology
• Prolactinomas are the commonest functioning pituitary tumour.
• Post-mortem studies show microadenomas in 10% of the population.
• During life, microprolactinomas are commoner than
macroprolactinomas, and there is a ♀ preponderance of
microprolactinomas.
Pathogenesis
Unknown. Occur in 20% of patients with MEN-1 (prolactinomas are the
commonest pituitary tumour in MEN-1 and may be more aggressive than
sporadic prolactinomas). Malignant prolactinomas are very rare and may
harbour RAS mutations.
Clinical features
Hyperprolactinaemia (microadenomas and macroadenomas)
• Galactorrhoea (up to 90% ♀, <10% ♂).
• Disturbed gonadal function in ♀ presents with menstrual disturbance
(up to 95%)—amenorrhoea, oligomenorrhoea, or with infertility and
reduced libido.
• Disturbed gonadal function in ♂ presents with loss of libido and/
or erectile dysfunction. Presentation with reduced fertility and
oligospermia or gynaecomastia is unusual.
• Hyperprolactinaemia is associated with a long-term risk of d BMD.
• Hyperprolactinaemia inhibits GnRH release, leading to d LH secretion.
There may be a direct action of PRL on the ovary to interfere with LH
and FSH signalling which inhibits oestradiol and progesterone secretion
and also follicle maturation.
Mass effects (macroadenomas only)
• Headaches and visual field defects (uni- or bitemporal field defects).
• Hypopituitarism.
• Invasion of the cavernous sinus may lead to cranial nerve palsies.
• Occasionally, very invasive tumours may erode bone and present with
a CSF leak or s meningitis.
For causes of hyperprolactinaemia, see Box 2.12.
PROLACTINOMAS 141
Investigations of prolactinomas
Serum PRL
• The differential diagnosis of elevated PRL is shown in Box 2.12. Note
that the stress of venepuncture may cause mild hyperprolactinaemia,
so 2–3 levels should be checked, preferably through an indwelling
cannula after 30min.
• Serum PRL <2,000mU/L is suggestive of a tumour—either a
microprolactinoma or a non-functioning macroadenoma compressing
the pituitary stalk, with loss of dopamine inhibitory tone to the
lactotroph and subsequent hyperprolactinaemia.
• Serum PRL >4,000mU/L is diagnostic of a macroprolactinoma.
• Hook effect. This occurs where the assay utilizes antibodies recognizing
two ends of the molecule. One is used to capture the molecule and
one to label it. If PRL levels are very high, it may be bound by one
antibody but not by the other. Thus, above a certain concentration,
the signal will reduce, rather than increase, and very high PRL levels
will be spuriously reported as normal or only slightly raised.
Thyroid function and renal function
Hypothyroidism and chronic renal failure are causes of hyperprolactinaemia.
Imaging
• MRI. Microadenomas usually appear as hypointense lesions within
the pituitary on T1-weighted images. Negative imaging is an indication
for contrast enhancement with gadolinium. Stalk deviation or gland
asymmetry may also suggest microadenoma.
• Macroadenomas are space-occupying tumours, often associated with
bony erosion and/or cavernous sinus invasion.
Macroprolactin (‘big’ PRL)
Occasionally, aggregate forms (150–170kDa) of PRL are detected in the
circulation. Although these are measurable in the prolactin assay, they
do not interfere with reproductive function but may be found in 10% of
patients referred. Typically, there is hyperprolactinaemia with regular ovu-
latory menstrual cycles. Assays for macroprolactin are available, using PEG
(polyethylene glycol) precipitation, where low recovery of PRL demon-
strates the presence of macroprolactin, or gel filtration chromatography
(gold standard).
‘IDIOPATHIC’ HYPERPROLACTINAEMIA 143
‘Idiopathic’ hyperprolactinaemia
When no cause is found following evaluation, as described on b p. 141,
the hyperprolactinaemia is designated idiopathic but, in many cases, is
likely to be due to a tiny microprolactinoma which is not demonstrable
on current imaging techniques. In other cases, it may be due to altera-
tions in hypothalamic regulation. Follow-up of these patients shows that,
in one-third, PRL levels return to normal; in 10–15%, there is a further
increase in PRL, and, in the remainder, PRL levels remain stable.
144 CHAPTER 2 Pituitary
Treatment of prolactinomas
Aims of therapy
• Microprolactinomas. Restoration of gonadal function.
• Macroprolactinomas.
• Reduction in tumour size and prevention of tumour expansion.
• Restoration of gonadal function.
• Although microprolactinomas may expand in size without treatment, the
vast majority do not. Therefore, although restoration of gonadal function
is usually achieved by lowering PRL levels, ensuring adequate sex
hormone replacement is an alternative if the tumour is monitored in size.
• Macroprolactinomas, however, will continue to expand and lead to
pressure effects. Definitive treatment of the tumour is, therefore,
necessary.
Drug therapy—dopamine agonists
• Dopamine agonist treatment (b see Dopamine agonists, p. 204)
leads to suppression of PRL in most patients, with s effects of
normalization of gonadal function and termination of galactorrhoea.
Tumour shrinkage occurs at a variable rate (from 24h to 6–12 months)
and extent and must be carefully monitored. Continued shrinkage
may occur for years. Slow chiasmal decompression will correct
visual field defect in the majority of patients, and immediate surgical
decompression is not necessary. Lack of improvement of visual fields,
despite tumour shrinkage, makes improvement with surgery unlikely.
Restoration of other hormonal axes may occur with tumour shrinkage.
• Cabergoline is more effective in normalization of PRL in
microprolactinoma (83% compared with 59% on bromocriptine), with
fewer side effects than bromocriptine.
• Although cabergoline in higher doses used for Parkinson’s disease can
cause right-sided cardiac fibrosis, there is no evidence for this using the
lower doses necessary for the control of PRL levels (see Box 2.13).
• Dopamine agonist resistance (see Box 2.14) may occur when there are
reduced numbers of D2 receptors.
• Tumour enlargement following initial shrinkage on treatment is usually
due to non-compliance. A rare possibility, however, is carcinoma.
Drug therapy—oestrogens
Oestrogen replacement, rather than dopamine agonist therapy, may be
appropriate in ♀ with idiopathic hyperprolactinaemia or microprolacti-
nomas where fertility and galactorrhoea are not issues. Small short-term
series suggest no evidence of tumour enlargement. However, individual
cases where tumour enlargement has occurred make monitoring of PRL
important.
Surgery
b see Transsphenoidal surgery, p. 196.
• Since the introduction of dopamine agonist treatment, transsphenoidal
surgery is indicated only for patients who are resistant to, or intolerant
of, dopamine agonist treatment. The cure rate for macroprolactinomas
TREATMENT OF PROLACTINOMAS 145
Further reading
Barber T, et al. (2011). Recurrence of hyperprolactinaemia following discontinuation of dopamine
agonist therapy in patients with prolactinoma occurs commonly especially in macroprolac-
toinoma. Clin Endocrinol 75, 819–24.
Bevan JS, Webster J, Burke CW, et al. (1992). Dopamine agonists and pituitary tumour shrinkage.
Endocrinol Rev 13, 220–40.
Casaneuva FF, Molitch ME, Schlechte JA, et al. (2006). Guidelines of the Pituitary Society for the
diagnosis and management of prolactinomas. Clin Endocrinol 65, 265–73.
Dekkers OM, Lagro J, Burman P, Jørgensen JO, Romijn JA, Pereira AM (2011). Recurrence of hyper-
prolactinaemia after withdrawal of dopamine agonists: systematic review and meta-analysis. J
Clin Endocrinol Metab 95, 43–51.
Karavitaki N, Thanabalasingham G, Shore HC (2006). Do the limits of serum prolactin in disconnec-
tion hyperprolactinaemia need re-definition? A study of 226 patients with histologically verified
non-functioning pituitary macroadenoma. Clin Endocrinol 65, 524–9.
Melmed S, et al. (2011). Diagnosis and treatment of hyperprolactinaemia: an Endocrine Society
Clinical practice guideline. J Clin Endocrinol Metab 96, 273–88.
Molitch ME (1985). Pregnancy and the hyperprolactinaemic woman. New Engl J Med 312, 1364–70.
Molitch ME (1992). Pathologic hyperprolactinaemia. Endocrinol Metabo Clin N Am 21, 877–910.
Molitch ME (2002). Medical management of prolactinomas. Pituitary 5, 55–65.
Molitch ME (2003). Dopamine resistance of prolactinomas. Pituitary 6, 19–27.
Molitch ME (2008). Drugs and prolactin. Pituitary 11, 209–18.
Suliman SG, Gurlek A, Byrne JV (2007). Non-surgical cerebrospinal fluid rhinorrhoea in invasive
macroprolactinoma: incidence, radiological and clinicopathological features. J Clin Endocrinol
Metab 92, 3829–35.
148 CHAPTER 2 Pituitary
Definition of acromegaly
Acromegaly is the clinical condition resulting from prolonged excessive
GH and hence IGF-1 secretion in adults. GH secretion is characterized by
blunting of pulsatile secretion and failure of GH to become undetectable
during the 24h day, unlike normal controls.
Epidemiology of acromegaly
• Rare. Equal sex distribution.
• Prevalence 40–86 cases/million population. Annual incidence of new
cases in the UK is 4/million population.
• Onset is insidious, and there is, therefore, often a considerable delay
between onset of clinical features and diagnosis. Most cases are
diagnosed at 40–60 years. Typically, acromegaly occurring in an older
patient is a milder disease, with lower GH levels and a smaller tumour.
Pituitary gigantism
The clinical syndrome resulting from excess GH secretion in children prior
to fusion of the epiphyses.
• Rare.
• i growth velocity without premature pubertal manifestations should
arouse suspicion of pituitary gigantism.
• Differential diagnosis. Marfan’s syndrome, neurofibromatosis,
precocious pubertal disorders, cerebral gigantism (large at birth with
accelerated linear growth and disproportionately large extremities—
associated normal IGF-1 and GH).
• Arm span > standing height is compatible with eunuchoid features and
suggests onset of disease before epiphyseal fusion (pituitary gigantism).
CAUSES OF ACROMEGALY 149
Causes of acromegaly
• Pituitary adenoma (>99% of cases). Macroadenomas 60–80%,
microadenomas 20–40%. Local invasion is common, but frank
carcinomas are very rare.
• GHRH secretion:
• Hypothalamic secretion.
• Ectopic GHRH, e.g. carcinoid tumour (pancreas, lung) or other
neuroendocrine tumours.
• Pituitary shows global enlargement. Somatroph hyperplasia seen on
histology.
• Ectopic GH secretion. Very rare (e.g. pancreatic islet cell tumour,
lymphoreticulosis).
• There has been some progress on the molecular pathogenesis of the
GH-secreting pituitary adenomas—as mutations of the Gsα are found
in up to 40% of tumours. This leads to an abnormality of the G protein
that usually inhibits GTPase activity in the somatotroph.
• Gene mutations have been shown in some young patients with
acromegaly who also have large tumours (familial isolated pituitary
adenoma) in whom a family history should be sought.
150 CHAPTER 2 Pituitary
Associations of acromegaly
• MEN-1. Less common than prolactinomas (b see MEN type 1,
p. 586).
• Carney complex. AD, spotty cutaneous pigmentation, cardiac and other
myxomas, and endocrine overactivity, particularly Cushing’s syndrome
due to nodular adrenal cortical hyperplasia and GH-secreting pituitary
tumours in <10% of cases. Mainly due to activating mutations of
protein kinase A (b see p. 583).
• McCune–Albright syndrome (b see p. 576). Caused by somatic
mosaicism for the gsp mutation.
• Familial isolated pituitary adenomas:
• Existence of two or more cases of acromegaly or gigantism in a
family that does not exhibit MEN-1 or Carney complex.
• Autosomal dominant.
• Most have acromegaly, but acromegaly and prolactinoma families
exist as rarely as do NFA families.
• 30–50% cases have a mutation in the AIP gene (tumour
suppressor gene).
• Early-onset disease (<30 years).
• Poor response to somatostatin analogues.
• Take a careful family history, especially in acromegalic patients with
a large pituitary tumour presenting below the age of 30 years, and
consider screening for AIP.
• See Box 2.25 for causes of macroglossia.
Investigations of acromegaly
Oral glucose tolerance test (OGTT)
• In acromegaly, there is failure to suppress GH to <0.33 micrograms/L
in response to a 75g oral glucose load. In contrast, the normal
response is GH suppression to undetectable levels.
• False +ves. Chronic renal and liver failure, malnutrition, diabetes mellitus,
heroin addiction, adolescence (due to high pubertal GH surges).
Random GH
Not useful in the diagnosis of acromegaly as, although normal healthy sub-
jects have undetectable GH levels throughout the day, there are pulses of
GH which are impossible to differentiate from the levels seen in acromeg-
aly. However, in untreated patients, a random GH <0.33 micrograms/L
practically excludes the diagnosis.
IGF-1
Useful in addition to the OGTT in differentiating patients with acromegaly
from normals, as it is almost invariably elevated in acromegaly, except in
severe intercurrent illness. It has a long half-life, as it is bound to binding
proteins, and reflects the effect of GH on tissues. However, abnormalities
of GH secretion may remain while IGF-1 is normal.
MRI
MRI usually demonstrates the tumour (98%) and whether there is extra-
sellar extension, either suprasellar or into the cavernous sinus.
Pituitary function testing
(b also see Insulin tolerance test (ITT), p. 114.) Serum PRL should be
measured, as some tumours co-secrete both GH and PRL.
Serum calcium
Some patients are hypercalciuric due to i 1,25-DHCC, as GH stimulates
renal 1α-hydroxylase. There may be an i likelihood of renal stones due
to hypercalcaemia as well as hypercalciuria (which occurs in 80%). Rarely,
hypercalcaemia may be due to associated MEN-1 and hyperparathyroidism.
GHRH
Occasionally, it is not possible to demonstrate a pituitary tumour, or the
pituitary gland MRI reveals global enlargement and histology reveals hyper-
plasia. A serum GHRH, in addition to radiology of the chest and abdomen,
may then be indicated to identify the cause, usually a GHRH-secreting
carcinoid of lung or pancreas.
INVESTIGATIONS OF ACROMEGALY 153
Management of acromegaly
The management strategy depends on the individual patient and also
on the tumour size. Lowering of GH is essential in all situations (see
Fig. 2.7).
Transsphenoidal surgery
(b also see Transsphenoidal surgery, p. 196.)
• This is usually the first line for treatment in most centres.
• Reported cure rates vary: 40–91% for microadenomas and 10–48% for
macroadenomas, depending on surgical expertise.
• A GHDC should be performed following surgery to assess whether
‘safe’ levels of GH and IGF-1 have been attained. If the mean GH is
<2.5 micrograms/L, then the patient can be followed up with annual
IGF-1 and/or GH assessment. If safe levels of GH have not been
achieved, then medical treatment and/or radiotherapy is indicated.
• Surgical debulking of a large macroadenoma should be undertaken,
even if cure is not expected, because this lowers GH levels and
improves the cure rate with subsequent somatostatin analogues.
Tumour recurrence following surgery
This is defined as tumour regrowth and increase in GH levels, lead-
ing to active acromegaly following post-operative normalization of GH
levels. Using the definition of post-operative cure as mean GH <2.5
micrograms/L, the reported recurrence rate is low (6% at 5 years).
Radiotherapy
(b also see Technique, p. 200.)
• This is usually reserved for patients following unsuccessful
transsphenoidal surgery, only occasionally is it used as p therapy. The
largest fall in GH occurs during the first 2 years, but GH continues to
fall after this. However, normalization of mean GH may take several
years and, during this time, adjunctive medical treatment (usually
with somatostatin analogues) is required. With a starting mean GH
>25 micrograms/L, it takes, on average, 6 years to achieve mean
GH <2.5 micrograms/L, compared with 4 years with a starting mean
GH <25 micrograms/L.
• After radiotherapy, somatostatin analogues should be withdrawn on
an annual basis to perform a GHDC to assess progress and identify
when mean GH <2.5 micrograms/L and, therefore, radiotherapy has
been effective and somatostatin analogue treatment is no longer
required.
• Radiotherapy can induce GH deficiency which may need GH therapy.
Definition of cure
• Controlled disease is, most recently, defined as a random GH <1
micrograms/L or nadir GH <0.4 micrograms/L on GTT + normal
age-related IGF-1.
MANAGEMENT OF ACROMEGALY 155
Microadenoma Macroadenoma
Surgery Surgery
Medical treatment
Somatostatin analogues
(60% normal IGF-1 and GH <2.5µg/L)
or
Dopamine agonists
(10–20% GH <2.5µg/L) IGF-I normal
or
GH receptor antagonists (IGF-1 normal >90%)
GH <2.5µg/L GH <2.5µg/L
IGF-1 normal IGF-1 high
Pathophysiology of Cushing’s
disease
• The vast majority of Cushing’s syndrome is due to a pituitary
ACTH-secreting corticotroph microadenoma. The underlying aetiology
is ill understood.
• Occasionally, corticotroph adenomas reach larger sizes
(macroadenomas) and rarely become invasive or malignant. The
tumours typically maintain some responsiveness to the usual feedback
control factors that influence the normal corticotroph (e.g. high doses
of glucocorticoids and CRH). However, this may be lost, and the
tumours become fully autonomous, particularly in Nelson’s syndrome.
• NB Crooke’s hyaline change is a fibrillary appearance seen in the
non-tumourous corticotroph associated with elevated cortisol levels
from any cause.
For causes of Cushing’s syndrome, see Box 2.27.
2000–4000
1000–2000
900
Plasma immunoreactive ACTH (ng/l)
700
500
300
250
200
150
100
50
70 18 81
Fig. 2.8 Plasma ACTH levels (9 a.m.) in patients with pituitary-dependent
Cushing’s disease, adrenal tumours, and ectopic ACTH secretion. From Besser M
and Thorner GM (1994). Clinical Endocrinology 2nd edn. Mosby.
166 CHAPTER 2 Pituitary
ectopic ACTH
1000
500
<5
−15 0 30 60 90 120
Time (h)
Fig. 2.9 CRH test in pituitary-dependent and ectopic disease. In the patient with
pituitary-dependent disease, the characteristic marked plasma cortisol rise after an
IV bolus of 100 micrograms of CRH is seen. Serum cortisol levels are unaltered
in the patient with ectopic ACTH secretion. Reproduced from Besser M and
Thorner GM (1994). Clinical Endocrinology, 2nd edn. Mosby. Copyright Elsevier, with
permission.
Pituitary imaging
MRI, following gadolinium enhancement which significantly increases
the pickup rate, localizes corticotroph adenomas in up to 80% of cases.
However, it should be remembered that at least 10% of the normal popu-
lation harbour microadenomas and, therefore, the biochemical investiga-
tion of these patients is essential, as a patient with an ectopic source to
Cushing’s syndrome may have a pituitary ‘incidentaloma’.
Other pituitary function
Hypercortisolism suppresses the thyroidal, gonadal, and GH axes, leading
to lowered levels of TSH and thyroid hormones as well as reduced gon-
adotrophins, gonadal steroids, and GH.
INVESTIGATIONS OF CUSHING’S DISEASE 167
Pituitary
Right Left
Adenoma
Fig. 2.10 Simultaneous bilateral inferior petrosal sinus and peripheral vein
sampling for ACTH. The ratio of >3 between the left central and peripheral
vein confirm a diagnosis of Cushing’s disease. Reproduced from Besser M and
Thorner GM (1994). Clinical Endocrinology, 2nd edn. Mosby. Copyright Elsevier, with
permission.
168 CHAPTER 2 Pituitary
Medical treatment
(See Table 2.10.)
• This is indicated during the preoperative preparation of patients
or while awaiting radiotherapy to be effective or if surgery or
radiotherapy are contraindicated.
• Inhibitors of steroidogenesis: metyrapone is usually used first-line,
but ketoconazole should be used as first-line in children, as it is
unassociated with i adrenal metabolites. There is also a suggestion
that ketoconazole may have a direct action on the corticotroph as well
as lowering cortisol secretion.
• Disadvantage of these agents inhibiting steroidogenesis is the need to
increase the dose to maintain control, as ACTH secretion will increase
as cortisol concentrations decrease.
• Steroidogenesis inhibitors may be used with glucocorticoid
replacement regimen to completely inhibit cortisol or with an aim for
partial inhibition of cortisol production.
• The dose of these drugs needs to be titrated against the cortisol
results from a day curve (cortisol taken at 9 a.m., 12 noon, 3 p.m.,
6 p.m.), aiming for a mean cortisol of 150–300nmol/L, as this
approximates the normal production rate.
• Response rates for drugs that reduce ACTH/CRH synthesis/release,
e.g. somatostatin agonists (pasireotide affecting SSTR5 and 2),
bromocriptine, valproate, and cyproheptadine, are variable. Pasireotide
improves urinary cortisol to normal in about 30%, but carbohydrate
tolerance may worsen.
• Successful treatment (surgery or radiotherapy) of Cushing’s disease
leads to cortisol deficiency and, therefore, glucocorticoid replacement
therapy is essential. In addition, patients who have undergone bilateral
adrenalectomy require fludrocortisone. These patients should all
receive instructions for intercurrent illness and carry a MedicAlert
bracelet and steroid card.
TREATMENT OF CUSHING’S DISEASE 171
Management
Surgery
(Aspects of pituitary surgery are also covered on b p. 196.)
• The initial definitive management in virtually every case is surgical.
This removes mass effects and may lead to some recovery of pituitary
function in around 10%. The majority of patients can be operated on
successfully via the transsphenoidal route.
• Close follow-up is necessary after surgery, as tumour regrowth can
only be detected using pituitary imaging and visual field assessment.
Radiotherapy
• The use of post-operative radiotherapy remains controversial. Some
centres advocate its use for every patient following surgery; others
reserve its use for those patients who have had particularly invasive
or aggressive tumours removed or those with a significant amount of
residual tumour remaining (e.g. in the cavernous sinus).
• The regrowth rate at 10 years without radiotherapy approaches
45%, and there are no good predictive factors for determining which
tumours will regrow. However, administration of post-operative
radiotherapy reduces this regrowth rate to <10%. As discussed in
b Complications, p. 76, however, there are sequelae to
radiotherapy—with a significant long-term risk of hypopituitarism and
a possible i risk of visual deterioration and malignancy in the field of
radiation.
Medical treatment
• Unlike the case for GH- and PRL-secreting tumours, medical therapy
for NFAs is usually unhelpful, although there have been reports of the
somatostatin agonist octreotide leading to tumour shrinkage and/or
visual field improvement in some cases.
• Hormone replacement therapy is required to treat any hypopituitarism
(b see p. 127).
• Visual field defects at diagnosis may improve following surgery in the
majority, and improvement may continue for a year following tumour
debulking.
Prediction of regrowth of NFAs?
No markers that provide certainty. However, the following have been
suggested as useful markers to raise suspicion of aggressive behaviour:
• Younger age.
• Preoperative cavernous sinus invasion and post-operative suprasellar
extension.
• Atypical features on histology (elevated mitotic index, MIB-1 labelling
index >3%, and macronucleoli).
Biochemical markers for NFAs?
• The majority of patients lack a hormone marker—despite
approximately half immunostaining positively for gonadotrophins and
containing secretory granules at the EM level.
• A minority of patients have elevated circulating FSH/LH levels (b see
Gonadotrophinomas, p. 178).
176 CHAPTER 2 Pituitary
Follow-up
(See Fig. 2.3.)
• Patients who have not received post-operative irradiation require
careful, long-term follow-up with serial pituitary imaging and visual
field assessment. The optimal protocol is still not known, but an
accepted practice is to image in the first 3 months following surgery,
and then reimage annually for 5 years, and biannually thereafter.
Tumour recurrence has been reported at up to 15 years following
surgery, and, therefore, follow-up needs to be long-term. The amount
of post-operative tumour remnant predicates recurrence risk. Thus,
with an empty sella post-operatively, the 10-year recurrence is around
6%. With an intrasellar remnant, it is 50% and with an extrasellar
remnant 90%.
• Patients who have received post-operative radiotherapy, require
follow-up with annual visual field assessment and imaging only if a
deterioration is noted.
• Dopamine agonists may decrease recurrences, but this needs a proper
prospective study.
Prognosis
Patients with NFAs have a good prognosis once the diagnosis and
appropriate treatment, including replacement of hormone deficiency,
is performed. The main concern is the risk of tumour regrowth, with
subsequent visual failure. As mentioned earlier, the administration of
radiotherapy, although not without potential complications itself, sig-
nificantly reduces this risk. Non-irradiated patients require very close
follow-up in order to detect regrowth and perform repeat surgery or
administer radiotherapy.
NON-FUNCTIONING PITUITARY TUMOURS 177
Further reading
Dekkers OM, Pereira AM, Romijn JA (2008). Treatment and follow-up of nonfunctioning pituitary
macroadenomas. J Clin Endocrinol Metab 93, 3717–26.
Karavitaki N, et al. (2007). What is the natural history of nonoperated nonfunctioning pituitary
adenomas? Clin Endocrinol (Oxf) 67, 938–43.
Karavitaki N, Thanabalasingham G, Shore HC, et al. (2006). Do the limits of serum prolactin in dis-
connection hyperprolactinaemia need re-definition? A study of 226 patients with histologically
verified non-functioning pituitary macroadenoma Clin Endocrinol (Oxf) 65, 524–9.
Reddy R, et al. (2011). Can we ever stop imaging in surgically treated and radiotherapy-naive
patients with non-functioning pituitary adenoma? Eur J Endocrinol 165, 739–74.
178 CHAPTER 2 Pituitary
Gonadotrophinomas
Background
These are tumours that arise from the gonadotroph cells of the pituitary
gland and produce FSH, LH, or the α subunit. They are often indistinguish-
able from other non-functioning pituitary adenomas, as they are usually
silent and unassociated with excess detectable secretion of LH and FSH,
although studies demonstrate gonadotrophin/α subunit secretion in vitro.
Occasionally, however, these tumours do produce detectable excess hor-
mone in vivo.
Clinical features
• Gonadotrophinomas present in the same manner as other
non-functioning pituitary tumours, with mass effects and
hypopituitarism (b see p. 122).
• The rare FSH-secreting gonadotrophinomas may lead to
macroorchidism in ♂.
• May cause ovarian hyperstimulation in premenopausal females.
Investigations
The secretion of FSH and LH from these tumours is usually undetect-
able in the plasma. Occasionally, elevated FSH and, more rarely, LH are
measured. This finding is often ignored, particularly in post-menopausal ♀.
Management
These tumours are managed as non-functioning tumours. The potential
advantage of FSH/LH secretion from a functioning gonadotrophinoma is
that it provides a biochemical marker of presence of tumour for follow-up.
GONADOTROPHINOMAS 179
180 CHAPTER 2 Pituitary
Thyrotrophinomas
Epidemiology
These are rare tumours, comprising approximately 1% of all pituitary
tumours. The diagnosis may be delayed because the significance of an
unsuppressed TSH in the presence of elevated free thyroid hormone
concentrations may be missed. Approximately one-third of cases in the
literature have received treatment directed at the thyroid in the form of
radioiodine treatment or surgery before diagnosis. Unlike p hyperthyroid-
ism, thyrotrophinomas are equally common in ♂ and ♀; 5% are associ-
ated with MEN-1.
Tumour biology and behaviour
• The majority are macroadenomas (90%) and secrete only TSH, often
with α subunit in addition, but some co-secrete GH (55%) and/or
PRL (15%).
• The pathogenesis of thyrotoxicosis in the presence of normal TSH
levels is poorly understood, but there are reports of secretion of
TSH with i bioactivity, possibly due to changes in post-translational
hormone glycosylation.
• The observation that prior thyroid ablation is associated with
deleterious effects on the size of the tumour suggests some feedback
control and is similar to the aggressive tumours seen in Nelson’s
syndrome after bilateral adrenalectomy has been performed for
Cushing’s disease. Thyrotropin-secreting pituitary carcinoma has been
very rarely reported.
• 5% are associated with MEN-1.
Clinical features
(b see p. 28.)
• Clinical features of hyperthyroidism are usually present but often milder
than expected, given the level of thyroid hormones. In mixed tumours,
hyperthyroidism may be overshadowed by features of acromegaly.
• Mass effects. Visual field defects and hypopituitarism.
Investigations
(b see also pp. 11, 125.)
• TSH is inappropriately normal or elevated. The range of TSH that has
been described is <1–568mU/L, and one-third of untreated patients
had TSH in the normal range. There is no correlation between TSH
and T4.
• Free thyroid hormones. Elevated in 65% of patients.
• α subunit (raised in 65%). Typically, patients have an iα subunit:TSH
molar ratio (>1) (81%).
• Other anterior pituitary hormone levels. PRL and/or GH may be elevated
in mixed tumours (an OGTT may be indicated to exclude acromegaly).
• SHBG. Elevated into the hyperthyroid range.
• TRH test. Absent TSH response to stimulation with TRH (useful to
differentiate TSH-secreting tumours from thyroid hormone resistance
where the TSH response is normal or exaggerated).
THYROTROPHINOMAS 181
Pituitary incidentalomas
Definition
The term incidentaloma refers to an incidentally detected lesion that is
unassociated with hormonal hyper- or hyposecretion and has a benign
natural history.
The increasingly frequent detection of these lesions with technologi-
cal improvements and more widespread use of sophisticated imaging has
led to a management challenge—which, if any, lesions need investigation
and/or treatment, and what is the optimal follow-up strategy (if required
at all)?
Epidemiology
• Autopsy studies have shown that 10–20% of pituitary glands
unsuspected of having pituitary disease harbour pituitary adenomas.
Approximately half the tumours stain for PRL, and the remainder
is –ve on immunostaining.
• Imaging studies using MRI demonstrate pituitary microadenomas in
approximately 10% of normal volunteers.
• Incidentally detected macroadenomas have been reported when
imaging has been performed for other reasons. However, these are
not true incidentalomas, as they are often associated with visual field
defects and/or hypopituitarism.
• Clinical significant pituitary tumours are present in about 1 in 1,000
patients.
Natural history
Incidentally detected microadenomas are very unlikely (<10%) to increase
in size whereas larger incidentally detected meso- and macroadenomas
are more likely (40–50%) to enlarge. Thus, conservative management in
selected patients may be appropriate for microadenomas which are inci-
dentally detected as long as careful follow-up imaging is in place and patients
are truly asymptomatic. Macroadenomas should be treated, if possible.
Clinical features
By definition, a patient with an incidentaloma should be asymptomatic.
Any patient who has an incidentally detected tumour should have visual
field assessment and a clinical review to ensure that this is not the initial
presentation of Cushing’s syndrome, acromegaly, or a prolactinoma.
Investigations
• Aims:
• Exclude any hormone hypersecretion from the tumour.
• Detect hypopituitarism.
• Investigation of hypersecretion of hormones should include measurement
of PRL, IGF-1 and an OGTT if acromegaly is suspected, 24h urinary free
cortisol and overnight dexamethasone suppression test, and thyroid
function tests (unsuppressed TSH in the presence of elevated T4).
• Others suggest that this approach is unnecessary, but, with limited data,
most endocrinologists would perform investigations as above.
PITUITARY INCIDENTALOMAS 183
Management
• All extrasellar macroadenomas (incidentally detected but, by
definition, not true incidentalomas) require definitive treatment.
• Tumours with excess hormone secretion require definitive treatment.
• Mass <1cm diameter—repeat MRI at 1, 2, and 5 years.
• Mass >1cm diameter—repeat MRI at 6 months, 1, 2, and 5 years.
Further reading
Fernando A, Karavitaki N, Wass JA (2010) Prevalence of pituitary adenomas: a community-based
cross-sectional study in Banbury, Oxfordshire (UK) Clin Endocrinol 72, 377–82.
Frida PU, et al. (2011). Pituitary incidentaloma: an Endocrine Society clinical practice guideline. Clin
Endocrinol Metab. 96, 894–904.
Karavitaki N, Collison K, Halliday J, et al. (2007). What is the natural history of nonoperated non-
functioning pituitary adenomas? Clin Endocrinol (Oxf) 67, 938–43.
Molitch ME (1997). Pituitary incidentalomas. End Met Clin North America 26, 725–40.
184 CHAPTER 2 Pituitary
Pituitary carcinoma
Definition
Pituitary carcinoma is defined as a p adenohypophyseal neoplasm with
craniospinal and/or distant systemic metastases (see Table 2.5 for types).
Epidemiology
These are extremely rare tumours, and only approximately 60 cases have
been reported in the world literature.
Pathology and pathogenesis
• The initial tumours and subsequent carcinomas show higher
proliferation indices than the majority of pituitary adenomas. They are
also likely to demonstrate p53 positivity and have an i mitotic index.
However, histology is unable to reliably distinguish between benign
invasive pituitary adenomas and carcinomas.
• The aetiology of these tumours is unknown, but the adenoma–
carcinoma sequence is followed in ACTH-secreting tumours: pituitary
adenoma causing Cushing’s disease, followed by locally invasive
adenoma (Nelson’s syndrome), leading to pituitary carcinoma.
• Metastatic spread outside the CNS is via lymphatic and vascular routes
while intra-CNS spread is via local invasion and tumour seeding.
Features
Virtually all pituitary carcinomas initially present as invasive pituitary mac-
roadenomas. After a variable interval of time (mean 6.5 years), the major-
ity presents with local recurrence. There is a tendency to systemic (liver,
lymph nodes, lungs, and bones), rather than craniospinal, metastases, but
metastases do not usually predominate in the clinical picture.
Neuroradiological classification of pituitary adenomas
(modified Hardy criteria)
• Grade 1—microadenoma.
• Grade 2—macroadenoma with or without suprasellar extension.
• Grade 3—locally invasive tumour with bony destruction and tumour in
the cavernous or sphenoid sinus.
• Grade 4—spread within the CNS or extracranial dissemination.
Grades 3 and 4 are termed ‘invasive’.
Treatment
Treatment involves surgery, radiotherapy, and medical treatment. As mass
effects often predominate, initial debulking surgery may provide relief. It
may need to be repeated to maintain local control. Some advocate a
transsphenoidal route as less likely to disseminate tumour.
Radiotherapy or medical treatment provides palliation only.
Radiotherapy has been reported to be successful, in some cases, in con-
trolling growth and occasionally leading to regression. Stereotactic radio-
surgery may play a role. Medical treatment with dopamine agonists for
malignant prolactinomas and acromegaly has been reported, with vary-
ing results. Many pituitary carcinomas are dedifferentiated and, therefore,
PITUITARY CARCINOMA 185
Pituitary metastases
Incidence
0.1–28% autopsy series; <1% found at transsphenoidal surgery.
Epidemiology
Equal sex distribution. Age >60 (occasionally younger).
Features
Diabetes insipidus in almost 100%. Symptomatic pituitary failure and cra-
nial nerve defects less common. Often difficult to differentiate neuroradio-
logically from other pituitary mass lesions (adenoma, cyst, or inflammatory
pituitary mass). Many do not have symptoms, as features of end-stage
malignancy predominate. Disconnection hyperprolactinaemia may be a
feature, and very rare cases of endocrine hyperfunction related to metas-
tasis within a primary adenoma have been reported (see Box 2.16).
Diagnosis
Histology is required to confirm.
Treatment
• Of p tumour where possible.
• Management of endocrine symptoms.
• Decompression may be indicated for visual field defects.
Prognosis
Mean survival 6–7 months.
Craniopharyngiomas and
perisellar cysts
(See Box 2.17 for Rathke’s cleft cysts.)
Prevalence and epidemiology
• 0.065/1,000.
• Any age; only 50% present in childhood (<16 years).
Pathology
• Tumour arising from squamous epithelial remnants of
craniopharyngeal duct.
• Histology may be either adamantinomatous or squamous papillary.
• Cyst formation and calcification are common.
• Benign tumour, although infiltrates surrounding structures.
Features
• Raised intracranial pressure.
• Visual disturbance.
• Hypothalamo–pituitary disturbance.
• Growth failure in children.
• Precocious puberty and tall stature are less common.
• Anterior and posterior pituitary failure, including DI.
• Weight gain.
Other perisellar cysts
• Arachnoid.
• Epidermoid.
• Dermoid.
Investigations
• MRI/CT (CT may be helpful to evaluate bony erosion).
• Visual field assessment.
• Anterior and posterior pituitary assessment (b see p. 114 and p. 212).
Management
(See Fig. 2.4.)
• Gross total removal is the aim of treatment, as this is associated with
a significantly lower recurrence rate. This may be via a transfrontal
or transsphenoidal route. If total removal cannot be safely achieved,
adjuvant radiotherapy is beneficial in reducing recurrence.
• Restoration of pituitary hormone deficiencies is extremely unlikely
following surgery.
• Cystic lesions may be treated with aspiration alone, although
radiotherapy reduces the likelihood of reaccumulation.
Prognosis
• Craniopharyngiomas are associated with d survival (up to 5x the
mortality of the general population).
• Recurrence following initial treatment may present early or several
decades following initial treatment. Childhood and adult onset lesions
behave similarly.
CRANIOPHARYNGIOMAS AND PERISELLAR CYSTS 189
Further reading
Karivitaki N, Cudlip S, Adams CB, et al. (2006). Craniopharyngiomas. Endocr Rev 27, 371–93.
Trifanescu R, et al. (2011). Outcome in surgically treated Rathke’s cleft cysts: long-term monitoring
needed. Eur J Endocrinol 165, 33–7.
Trifanescu R, et al. (2012). Rathke’s cleft cysts. Clin Endocrinol (Oxf) 76, 51–60.
190 CHAPTER 2 Pituitary
Parasellar tumours
Meningiomas
• Suprasellar meningiomas arise from the tuberculum sellae or the
chiasmal sulcus.
• Usually present with a chiasmal syndrome where loss of visual acuity
occurs in one eye, followed by reduced acuity in the other eye.
• Differentiation from a p pituitary tumour can be difficult where there
is downward extension into the sella.
• MRI is the imaging of choice. T1-weighted images demonstrate
meningiomas as isodense with grey matter and hypointense with
respect to pituitary tissue, with marked enhancement after gadolinium.
• Cerebral angiography also demonstrates a tumour blush.
• Management is surgical and may also be complicated by haemorrhage,
as these are often very vascular tumours. They are relatively
radioresistant, but inoperable or partially removed tumours may
respond. As they are slow-growing, a conservative approach with
regular imaging may be appropriate.
• Associations include type 2 neurofibromatosis (b p. 578).
Clivus chordomas
• Rare. Arise from embryonic crest cells of the notochord.
• May present with cranial nerve palsies (III, VI, IX, X) or pyramidal tract
dysfunction.
• Anterior and posterior pituitary hypofunction is reported.
• Often invasive and relentlessly progressive.
• Treatment is surgical, followed by radiotherapy in some cases,
although they are relatively radioresistant. Data on radiosurgery are
not yet available, but this may be considered.
Hamartomas
• Non-neoplastic overgrowth of neurones and glial cells.
• Rare. May present with seizures—typically gelastic (laughing).
• May release GnRH leading to precocious puberty or, very rarely,
GHRH leading to disorders of growth or acromegaly.
• Appear as homogeneous, isointense with grey matter, pedunculated or
sessile non-enhancing tumours on T1-weighted MRI scans.
Management
Tumours do not enlarge, and, therefore, treatment is of endocrine conse-
quences—most commonly, precocious puberty.
Ependymomas
• Intracranial ependymomas typically affect children and adolescents.
• Pituitary insufficiency may follow craniospinal irradiation.
• Occasionally, third ventricle tumours may interfere with hypothalamic
function.
Further reading
Whittle IR, Smith C, Navoo P, et al. (2004). Meningiomas. Lancet 363, 1535–43.
PARASELLAR TUMOURS 191
192 CHAPTER 2 Pituitary
Tuberculosis
TB may present as a tuberculoma which may compromise hypothalamic
or pituitary function. DI is common. Most patients have signs of TB else-
where but not invariably so. Transsphenoidal biopsy is, therefore, some-
times required. An alternative strategy is antituberculous treatment with
empirical glucocorticoid treatment.
Further reading
Freda PU, Post KD (1999). Differential diagnosis of sellar masses. Endocrinol Metabol Clin N Am
28, 81.
194 CHAPTER 2 Pituitary
Lymphocytic hypophysitis
Background
This is a rare inflammatory condition of the pituitary.
Epidemiology
Lymphocytic hypophysitis occurs more commonly in ♀ and usually pre-
sents during late pregnancy or the first year thereafter.
Pathogenesis
Ill understood—probably autoimmune. Approximately 25% of cases of
lymphocytic hypophysitis have been associated with other autoimmune
conditions—Hashimoto’s thyroiditis in the majority but also pernicious
anaemia.
There is a recent association with ipilimumab, immunostimulating agent
a monoclonal antibody against cytotoxic T lymphocyte antigen 4 (CTLA4).
Pathology
• Somatotroph and gonadotroph function are more likely to be
preserved than corticotroph or thyrotroph function, unlike the findings
in hypopituitarism due to a pituitary tumour. The posterior pituitary
is characteristically spared so that DI is not part of the picture, but
there are occasional reports of coexistent or isolated DI, presumably
because of different antigens.
• Lymphocytic hypophysitis has occasionally involved the cavernous
sinus and extraocular muscles.
• Light microscopy typically reveals a lymphoplasmacytic infiltrate,
occasionally forming lymphoid follicles, with variable destruction of
parenchyma and fibrosis.
Clinical features
• Mass effects, leading to headache and visual field defects.
• Often a temporal association with pregnancy.
• Hypopituitarism (ACTH and TSH deficiency, less commonly
gonadotrophin and GH deficiency).
• Posterior pituitary involvement and cavernous sinus involvement occur
less commonly.
• See Box 2.18 for classification.
Investigations
• Investigation of hypopituitarism is essential and may not be thought
of because gonadotrophin secretion often remains intact, leaving the
potentially life-threatening ACTH deficiency unsuspected.
• MRI shows an enhancing mass, with variably loss of hyperintense bright
spot of neurohypophysis, thickening of pituitary stalk, and enlargement
of the neurohypophysis. Suprasellar extension often appears
tongue-like along the pituitary stalk. There may be central necrosis but
no calcification.
• Biopsy of the lesion is often required but may be avoided in the
presence of typical features.
LYMPHOCYTIC HYPOPHYSITIS 195
Complications
(See also Box 2.20 and Table 2.6.)
• Patients should be informed about the possible complications
of transsphenoidal surgery prior to consent. The commonest
complications are DI, which may be transient or permanent (5% and
0.1%, respectively; often higher in Cushing’s disease and prolactinoma),
and the development of new anterior pituitary hormonal deficiencies
(uncommon with microadenomas; approximately 10% of TSA for
macroadenomas).
• Other complications include meningitis, CSF leak, visual deterioration,
haemorrhage (rare), and transient hyponatraemia, usually 7 days
post-operatively.
Transfrontal craniotomy
Indications
• Pituitary tumours with major suprasellar and lateral invasion where
transsphenoidal surgery is unlikely to remove a significant proportion
of the tumour.
• Parasellar tumours, e.g. meningioma.
Complications
• In addition to the complications of transsphenoidal surgery, brain
retraction can lead to cerebral oedema or haemorrhage.
• Manipulation of the optic chiasm may lead to visual deterioration.
• Vascular damage.
• Damage to the olfactory nerve.
Perioperative management
Similar to that for patients undergoing transsphenoidal surgery (b see
Transsphenoidal surgery, p. 196).
Post-operative management
• Recovery is typically slower than after transsphenoidal surgery.
• Prophylactic anticonvulsants are administered for up to 1 year.
• The DVLA must be advised of surgery and relevant regulations to be
followed.
Further reading
Laws ER, Thapar K (1999). Pituitary surgery. Endocrinol Metab Clin N Am 28, 119.
200 CHAPTER 2 Pituitary
Pituitary radiotherapy
Indications
(b see Box 2.21.)
• Pituitary radiotherapy is an effective treatment used to reduce the
likelihood of tumour regrowth following surgery, to further shrink a
tumour, and to treat persistent hormone hypersecretion (usually after
surgical resection or non-successful medical treatment).
• Pituitary radiotherapy is usually only administrable once in a lifetime.
Technique
(See Box 2.22 for focal forms of radiotherapy.)
• Conventional external beam 3-field radiotherapy is able to deliver a
beam of ionizing irradiation accurately to the pituitary fossa.
• Accurate targeting requires head fixation in a moulded plastic shell
to keep the head immobilized. The fields of irradiation are based on
simulation using MRI or CT scanning, and the volume is usually the
tumour margins plus 0.5cm in all planes. The preoperative tumour
volume is used for planning whereas the post-drug (dopamine agonist)
shrinkage films are used for prolactinomas. There are three portals—
two temporal and one anterior.
• The standard dose is 4500cGy in 25 fractions over 35 days, but 5,000
cGy may be used for relatively ‘radioresistant’ tumours, such as
craniopharyngiomas.
Efficacy
Radiotherapy is effective in reducing the chance of pituitary tumour
regrowth. Comparison of non-functioning tumour recurrence following
surgery and radiotherapy compared with surgery alone shows that radio-
therapy is effective in reducing the likelihood of regrowth (see Fig. 2.5).
80
radiotherapy
70
60
50 No
40 radiotherapy
30
20
10
0
10 15
Years after surgery
Fig. 2.5 Recurrence rates following radiotherapy. Modified from Gittoes NJL,
Bates AS, Tse W, et al. (1998). Radiotherapy for non-functioning pituitary tumours.
Clin Endocrinol 48, 331–7. With permission from Wiley Blackwell.
202 CHAPTER 2 Pituitary
Complications of pituitary
radiotherapy
Short term
• Nausea.
• Headache.
• Temporary hair loss at radiotherapy portals of entry.
Hypopituitarism (see Fig. 2.6)
• Anterior pituitary hormone deficiency occurs due to the effect of
irradiation on the normal pituitary or the hypothalamus, leading to
reduced hypothalamic-releasing hormone secretion. The total dose of
irradiation is one of the main determinants of the speed, incidence, and
extent of hypopituitarism (see Table 2.7).
• The onset of hypopituitarism is gradual, and the order of development
of deficiency is as for any other cause of developing hypopituitarism—
namely, GH first, followed by gonadotrophin and ACTH, followed
finally by TSH. Posterior pituitary deficiencies are very rare, but s
temporary mild hyperprolactinaemia may be seen after about 2 years
which gradually returns to normal.
Visual impairment
• The optic chiasm is particularly radioresistant but may undergo
damage thought to be due to vascular damage to the blood supply.
Visual deterioration typically occurs within 3 years of irradiation and is
progressive.
• The literature suggests that the risk is greatest with high total and daily
doses. A standard total dose of 4,500cGy and daily dose of 180cGy
appear to pose very little, if any, risk to the chiasm.
• Our practice is to avoid administration of radiotherapy, where
possible, when the chiasm is under pressure from residual tumour.
Radiation oncogenesis
• There is controversy as to whether pituitary irradiation leads to
the development of second tumours. There have been reports
of sarcomas, gliomas, and meningiomas developing in the field of
irradiation after 10–20 years. However, there are also reports of
gliomas and meningiomas occurring in non-irradiated patients with
pituitary adenomas. A retrospective review of a large series of patients
given pituitary irradiation suggested a risk of second tumour of 1.9%
by 20 years after irradiation when compared to the normal population
(but not patients with pituitary tumours).
• Subtle changes in neurocognition have also been suggested.
COMPLICATIONS OF PITUITARY RADIOTHERAPY 203
1.0
Probability of normal axis
TSH
0.5
LH/FSH
ACTH
GH
0
0 3 4 6 8 10
Years after treatment
Further reading
Brada M, Ford D, Ashley S, et al. (1992). Risk of second brain tumour after conservative surgery and
radiotherapy for pituitary adenoma. BMJ 304, 1343–6.
Jackson IMD, Noren G (1999). Role of gamma knife therapy in the management of pituitary
tumours. Endocrinol Metab Clin N Am 28, 133.
Jones A (1991). Radiation oncogenesis in relation to treatment of pituitary tumours. Clin Endocrinol
35, 379.
Loeffler JS, Shih HA (2011). Radiation therapy in the management of pituitary adenomas. J Clin
Endocrinol Metab 96, 1992–2003.
Minniti G, et al. (2005). Risk of second brain tumor after conservative surgery and radiotherapy
for pituitary adenoma: update after an additional 10 years. J Clin Endocrinol Metab 90, 800–4.
Plowman PN (1999). Pituitary adenoma radiotherapy. Clin Endocrinol 51, 265–71.
204 CHAPTER 2 Pituitary
Side effects
• Gallstones. At least 20–30% of patients develop gallstones or sludge on
octreotide (thought, by most, to antedate stone formation), but only
1%/year develop symptoms. The incidence is unknown on octreotide
LAR® or lanreotide. Symptoms may particularly occur if somatostatin
analogue therapy is withdrawn.
• GI due to inhibition of motor activity and secretion, leading to nausea,
abdominal cramps, and mild steatorrhoea. These usually settle
with time.
• Injection site pain obviated by allowing vial to warm to room
temperature before injecting.
• Hair loss (<10%).
Growth hormone receptor antagonist
Pegvisomant—newly developed treatment for acromegaly.
Mechanism of action
• Binds to GH receptor and induces internalization but blocks receptor
signalling, leading to reduction in IGF-1 (but not GH) production.
• Studies suggest that it is the most potent available medical therapy.
Normalization of IGF-1 in >90% treated patients.
Usage
• May be used with somatostatin analogues to decrease frequency of
injections.
Problems
• High cost.
• Further data required on the effects on pituitary tumour growth.
• Occasional deterioration in liver biochemistry.
• IGF-1 used to monitor effectiveness of treatment.
Temozolomide
• Is an alkylating chemotherapeutic agent effective in glioblastoma and
neuroendocrine tumours.
• It has been successfully used to treat pituitary carcinoma and invasive
and aggressive pituitary adenomas.
• Response may be predicated by O-6-methylguanine DNA
methyltransferase (MGMT) staining, which is lower in responders.
Further reading
McCormack AI, et al. (2011). Aggressive pituitary tumours: the role of temozolomide and the
assessment of MGMT status. Eur J Clin Invest 41, 1133–48.
Sandret L, Maison P, Chanson P (2011). Place of cabergoline in acromegaly: a meta-analysis. J Clin
Endocrinol Metab 96, 1327–35.
Sherlock M, Woods C, Sheppard MC (2011). Medical therapy in acromegaly. Nat Rev Endocrinol
7, 291–300.
Trainer PJ, et al. (2000). Treatment of acromegaly with the growth hormone–receptor antagonist
pegvisomant. N Engl J Med 342, 1171–7.
DRUG TREATMENT OF PITUITARY TUMOURS 207
208 CHAPTER 2 Pituitary
Posterior pituitary
Physiology and pathology
The posterior lobe of the pituitary gland arises from the forebrain and
comprises up to 25% of the normal adult pituitary gland. It produces
arginine vasopressin and oxytocin. Both hormones are synthesized in
the hypothalamic neurons of the supraoptic and paraventricular nuclei
and migrate as neurosecretory granules to the posterior pituitary before
release into the circulation. The hormones are unbound in the circulation,
and their half-life is short.
Oxytocin
• Oxytocin has no known role in ♂. It may aid contraction of the
seminal vesicles.
• In ♀, oxytocin contracts the pregnant uterus and also causes breast
duct smooth muscle contraction, leading to breast milk ejection during
breastfeeding. Oxytocin is released in response to suckling and also to
cervical dilatation during parturition. However, oxytocin deficiency has
no known adverse effect on parturition or breastfeeding.
• There are several, as yet, ill-understood features of oxytocin
physiology. For example, osmotic stimulation may also lead to
oxytocin secretion. Oxytocin may play a role in the ovary and testis,
as ovarian luteal cells and testicular cells have both been shown to
synthesize it, although its subsequent role is not known.
Vasopressin and neurophysin
• Arginine vasopressin is the major determinant of renal water excretion
and, therefore, fluid balance. Its main action is to reduce free water
clearance.
• Vasopressin is a nonapeptide and derives from a large precursor
with a signal peptide and a neurophysin. The vasopressin gene is
located on chromosome 20 and is closely linked to the oxytocin gene.
Vasopressin travels to the posterior pituitary and undergoes cleavage
as it travels. Neurophysin is released with vasopressin but has no
further role after acting as a carrier protein in the neurons.
• Release of vasopressin occurs in response to changes in osmolality
detected by osmoreceptors in the hypothalamus. Large changes
in blood volume (5–10%) also influence vasopressin secretion.
Many substances modulate vasopressin secretion, including the
catecholamines and opioids.
• The main site of action of vasopressin is in the collecting duct and the
thick ascending loop of Henle where it increases water permeability
so that solute-free water may pass along an osmotic gradient to the
interstitial medulla. Vasopressin in higher concentrations has a pressor
effect. It also acts as an ACTH secretagogue synergistically with CRH.
POSTERIOR PITUITARY 209
210 CHAPTER 2 Pituitary
Investigations of DI
Diagnosis of type of DI
• Confirm large urine output (>3L/day).
• Exclude diabetes mellitus and renal failure (osmotic diuresis).
• Check electrolytes. Hypokalaemia and hypercalcaemia
(nephrogenic DI).
• Fluid deprivation test (see Box 2.31) and assessment of response to
vasopressin.
• Occasionally, further investigations are required, particularly when only
partial forms of the condition are present.
• Measurement of plasma vasopressin, osmolality, and thirst threshold:
• In response to infusion of 0.05mL/kg/min 5% hypertonic saline for
2h for cranial DI (no i vasopressin).
• In response to fluid deprivation for nephrogenic DI (vasopressin
levels rise, with no i urine osmolality).
• An alternative is a therapeutic trial of desmopressin, with monitoring
of sodium and osmolality.
Investigation of the cause
• MRI head:
• Looking for tumours (e.g. hypothalamic, pineal, or infiltration).
• May demonstrate loss of bright spot of posterior pituitary gland.
• Serum ACE (sarcoidosis) and tumour markers, e.g. BhCG (pineal
germinoma).
• Differential diagnosis of pituitary stalk lesions. Includes congenital,
inflammatory (sarcoid, langerhans, lymphocytic hypophysitis,
Wegener’s) and neoplastic (craniopharyngioma, metastases,
germinoma).
INVESTIGATIONS OF DI 213
Treatment of DI
Maintenance of adequate fluid input
In patients with partial DI and an intact thirst mechanism, drug therapy
may not be necessary if the polyuria is mild (<4L/24h).
Drug therapy
Desmopressin
• Vasopressin analogue, acting predominantly on the V2 receptors in the
kidney, with little action on the V1 receptors of blood vessels. It thus
has reduced pressor activity and i antidiuretic efficacy, in addition to a
longer half-life than the native hormone.
• Drug may be administered in divided doses, orally (100–1,000
micrograms/day), intranasally (10–40 micrograms/day), parenterally
(SC, IV, IM) (0.1–2 micrograms/day) or buccally. There is wide
variation in the dose required by an individual patient.
• Monitoring of serum sodium and osmolality is essential, as
hyponatraemia or hypo-osmolality may develop.
Lysine vasopressin
• The antidiuretic hormone of the pig family.
• Not used very often, as its effects are short-lived (1–3h) and it may
retain pressor activity. Intranasal administration. Dose 5–20U/day.
Chlorpropamide (100–500mg/day) and carbamazepine enhance the action
of vasopressin on the collecting duct.
Nephrogenic DI
• Correction of underlying cause (metabolic or drugs).
• High doses of desmopressin (e.g. up to 5 micrograms IM) can be
effective.
• Maintenance of adequate fluid input.
• Thiazide diuretics and prostaglandin synthase inhibitors (decrease the
action of prostaglandins which locally inhibit the action of vasopressin
in the kidney), e.g. indometacin, can be helpful.
Polydipsic polyuria
Management is difficult. Treatment of any underlying psychiatric disorder
is important.
Further reading
Fenske W, Allolio B (2012). Clinical review: Current state and future perspectives in the diagnosis
of diabetes insipidus: a clinical review. J Clin Endocrinol Metab 97, 3426–37.
Loh JA, Verbalis JG. (2008). Disorders of water and salt metabolism associated with pituitary
disease. Endocrinol Metab Clin North Am 37, 213–34.
Turcu AF, Erickson BJ, Lin E et al. (2013). Pituitary stalk lesions: The Mayo Clinic Experience. J Clin
Endocrinol Metab 98(5):1812–18.
TREATMENT OF DI 215
216 CHAPTER 2 Pituitary
Hyponatraemia
(See Table 2.12; for causes, see Box 2.32.)
Incidence
• 1–6% hospital admissions Na <130mmol/L.
• 15–22% hospital admissions Na <135mmol/L.
Features
• Depend on the underlying cause and also on the rate of development
of hyponatraemia. May develop once sodium reaches 115mmol/L or
earlier if the fall is rapid. Level at 100mmol/L or less is life-threatening.
• Features of excess water are mainly neurological because of brain
injury and depend on the age of the patient and rate of development.
They include confusion and headache, progressing to seizures and
coma. In hypervolaemic forms of excess water (where the fluid is
confined to the ECF, e.g. cardiac failure, nephrotic syndrome, and
cirrhosis), oedema and fluid overload are apparent. In contrast, in
the syndromes of excess water associated with SIADH, the fluid is
distributed throughout the ECF and ICF and there is no apparent fluid
overload. Salt deficiency presents with features of hypovolaemia, with
tachycardia and postural hypotension.
Investigations
(See Table 2.12.)
• Urinary sodium is very helpful in differentiating the underlying cause.
• Assess volume status.
• Other investigations as indicated, e.g. serum and urine osmolality,
cortisol, thyroid function, liver biochemistry, serum electrophoresis.
Treatment
Salt deficiency (renal or non-renal)
• Increase dietary salt.
• May need IV normal saline if dehydrated.
Excess water (cirrhosis, nephrotic syndrome, CCF)
• Fluid restriction to 500–750mL/24h.
• Occasionally, hypertonic (3%) saline (513mmol/L) may be required
in patients with acute symptomatic hyponatraemia. The appropriate
infusion rate can be calculated from the formula:
Rate of sodium replacement (mmol/h) = total body water (60% of body
weight) × desired correction rate (0.5–1mmol/h).
For example, for a 70kg ♂:
Na+ = 60% × 70kg × 1mmol/h = 42mmol/h
1000
= 42 × = 82mL / h of 3% saline
513
• Rapid normalization (faster than 0.5mmol/h) of sodium may be
associated with central pontine myelinolysis.
SIADH
b see Syndrome of inappropriate ADH (SIADH), p. 220.
HYPONATRAEMIA 219
Neurosurgical hyponatraemia
• Injudicious fluids.
• Diuretics.
• Drugs, e.g. carbamazepine, opiates.
• SIADH.
• Glucocorticoid deficiency.
• Cerebral salt wasting.
Unexplained hyponatraemia
• Common in elderly (i ADH release in response to i osmolality and
less effective suppression of ADH).
• i risk of hyponatraemia with SSRIs.
220 CHAPTER 2 Pituitary
Eating disorders
Anorexia nervosa
Features
• Typical presentation is a ♀ aged <25 with weight loss, amenorrhoea,
and behavioural changes.
• There is a long-term risk of severe osteoporosis associated with
>6 months of amenorrhoea. There is loss of bone mineral content
and bone density, with little or no recovery after resolution of the
amenorrhoea. Bone loss occurs at 2.5% per annum.
Endocrine abnormalities
• Deficiency of GnRH, low LH and FSH, normal PRL, and low oestrogen
in ♀ or testosterone in ♂.
• Elevated circulating cortisol (usually non-suppressible with
dexamethasone).
• Low normal thyroxine, reduced T3, and normal TSH.
• Elevated resting GH levels.
• In addition, it is common to find various metabolic abnormalities,
such as reduced magnesium, zinc, phosphorus, and calcium levels, in
addition to hyponatraemia, hypoglycaemia, and hypokalaemia.
• Weight gain leads to a reversion of the prepubertal LH secretory
pattern to the adult-like secretion. Administration of GnRH in a
pulsatile pattern leads to normalization of the pituitary–gonadal axis,
demonstrating that the p abnormality is hypothalamic.
Management
• The long-term treatment of these patients involves treatment of the
underlying condition and then management of osteoporosis, although
many patients will refuse oestrogen replacement.
• Resumption of menstrual function is important for recovery of spine
bone mineral density (BMD). Weight gain is important for hip BMD
recovery. Oral contraceptives do not help BMD recovery.
Bulimia
Features
• Typically occurs in ♀ who are slightly older than the group with
anorexia nervosa. Weight may be normal, and patients often deny the
abnormal eating behaviour. Patients gorge themselves, using artificial
means of avoiding excessive weight gain (laxatives, diuretic abuse,
vomiting). This may be a cause of ‘occult’ hypokalaemia.
• These patients may, or may not, have menstrual irregularity. If
menstrual irregularity is present, this is often associated with
inadequate oestrogen secretion and anovulation.
Further reading
Miller KK, Lee EE, Lawson EA, et al. (2006). Determinants of skeletal loss and recovery in anorexia
nervosa. J Clin Endocrinol Metab 91, 2931–7.
HYPOTHALAMUS 223
Hypothalamus
Pathophysiology
• The hypothalamus releases hormones that act as releasing hormones
at the anterior pituitary gland. It produces dopamine that inhibits PRL
release from the anterior pituitary gland. It also synthesizes arginine
vasopressin and oxytocin (b see p. 208).
• The commonest syndrome to be associated with the hypothalamus
is abnormal GnRH secretion, leading to reduced gonadotrophin
secretion and hypogonadism. Common causes are stress, weight loss,
and excessive exercise. Management involves treatment of the cause,
if possible. Administration of pulsatile GnRH is usually effective at
reversing the abnormality but rarely undertaken.
Hypothalamic syndrome
Uncommon but may occur due to a large tumour, following surgery for a
craniopharyngioma, or due to infiltration from, for example, Langerhans
cell histiocytosis. The typical features are hyperphagia and weight gain, loss
of thirst sensation, DI, somnolence, problems with temperature control,
and behaviour change. Management can be challenging, e.g. DI with the
loss of thirst sensation may require the prescription of regular fluid, in
addition to desmopressin, and monitoring with daily weights and fluid bal-
ance as a routine. See Box 2.35 for metabolic effects.
Pineal gland
Physiology
• The pineal gland lies behind the third ventricle, is highly vascular, and
produces melatonin and other peptides.
• During daylight, light-induced hyperpolarization of the retinal
photoreceptors inhibits signalling and the pineal gland is quiescent.
• Darkness stimulates the gland, and there is synthesis and release of
melatonin.
• Melatonin levels are usually low during the day, rise during the evening,
peak at midnight, and then decline independent of sleep.
• Exposure to darkness during the day does not increase melatonin
secretion.
Jet lag
• It is claimed that pharmacological doses of melatonin can reset the
body clock.
• Studies have suggested that melatonin may reduce the severity or
duration of jet lag. There are few safety data, and currently melatonin
does not have a product licence in the UK.
Pineal and intracranial germ cell tumours
• Incidence. 0.5–3% intracranial neoplasms.
• Epidemiology.♂ > ♀. All ages. Two-thirds present between age 10 and
21 years.
• Germ cell tumours are now classified as germinomas or
non-germinomatous germ cell tumours (choriocarcinoma, teratoma).
The latter term includes the tumours previously called ‘true’ pinealomas.
Features
• Raised intracranial pressure (compression of cerebral aqueduct),
headache, and lethargy.
• Visual disturbance (pressure on quadrigeminal plate).
• Parinaud’s syndrome (paralysis of upward gaze, convergent nystagmus,
Argyll–Robertson pupils).
• Ataxia.
• Pyramidal signs.
• Hypopituitarism.
• DI.
• Sexual precocity in boys (excess hCG secretion mimics LH, leading to
Leydig cell testosterone secretion or related to space-occupying mass).
Investigations
• Imaging of craniospinal axis MRI/CT (may detect second tumour/
metastases).
• CSF examination.
• Tumour markers (hCG, alpha fetoprotein).
• Cytology.
• Biopsy if tumour markers unhelpful.
• Pituitary function assessment.
PINEAL GLAND 225
Management
• External beam radiotherapy—standard therapy is to irradiate the
whole craniospinal axis.
• Germinomas are exquisitely radiosensitive.
• Chemotherapy may be indicated, e.g. vincristine, etoposide,
carboplatin.
• Craniospinal irradiation cures the majority of patients (90%).
Prognosis
Excellent for germinomas (70–85% 5-year survival), but non-germinomatous
germ cell tumours have a worse prognosis (15–40% 5-year survival).
Further reading
Hussaini M, et al. (2009). Pineal gland tumors: experience from the SEER database. J Neuro Oncol
94, 351–8.
Chapter 3 227
Adrenal
Anatomy 228
Physiology 230
Imaging 232
Mineralocorticoid excess: definitions 234
Primary aldosteronism 236
Conn’s syndrome—aldosterone-producing adenoma 238
Bilateral adrenal hyperplasia (bilateral idiopathic
hyperaldosteronism) 239
Glucocorticoid-remediable aldosteronism (GRA) 240
Aldosterone-producing carcinoma 241
Screening 242
Localization and confirmation of differential diagnosis 244
Treatment 246
Excess other mineralocorticoids 248
Adrenal Cushing’s syndrome 250
Treatment of adrenal Cushing’s syndrome 252
Adrenal surgery 256
Renal tubular abnormalities 258
Liddle’s syndrome 258
Bartter’s syndrome 259
Gitelman’s syndrome 260
Mineralocorticoid deficiency 262
Adrenal insufficiency 264
Addison’s disease 266
Autoimmune adrenalitis 268
Autoimmune polyglandular syndrome (APS) type 1 270
APS type 2 271
Investigation of primary adrenal insufficiency 272
Treatment of primary adrenal insufficiency 276
Long-term glucocorticoid administration 280
Adrenal incidentalomas 282
Phaeochromocytomas and paragangliomas 284
Investigations of phaeochromocytomas and paragangliomas 288
Screening for associated conditions 292
Management 294
228 CHAPTER 3 Adrenal
Anatomy
The normal adrenal glands weigh 4–5g. The cortex represents 90% of the
normal gland and surrounds the medulla. The arterial blood supply arises
from the renal arteries, aorta, and inferior phrenic artery. Venous drainage
occurs via the central vein into the inferior vena cava on the right and into
the left renal vein on the left. See Figs. 3.1 and 3.2.
Hypothalamus Aldosterone
ACTH
Glomerulosa cells
Angiotensin III ?
Adrenal
Angiotensin II
Converting
enzyme
(ACE)
Lungs
Angiotensin I
Renin
Angiotensinogen
Juxtaglomerular
cells
Sympathetic input
Blood pressure
Na
Kidney
Physiology
Glucocorticoids
Glucocorticoid (cortisol 10–20mg/day) (see Table 3.1) production occurs
from the zona fasciculata, and adrenal androgens arise from the zona retic-
ularis. Both of these are under the control of ACTH, which regulates both
steroid synthesis and also adrenocortical growth.
Mineralocorticoids
Mineralocorticoid (aldosterone 100–150 micrograms/day) (see Table 3.1)
synthesis occurs in zona glomerulosa, predominantly under the control of
the renin–angiotensin system (see Fig. 3.2), although ACTH also contrib-
utes to its regulation.
Androgens
The adrenal gland (zona reticularis and zona fasciculata) also produces sex
steroids in the form of dehydroepiandrostenedione (DHEA) and andros-
tenedione. The synthetic pathway is under the control of ACTH.
Urinary steroid profiling provides quantitative information on the bio-
synthetic and catabolic pathways. Profiling can be useful in:
• Mineralocorticoid hypertension.
• Polycystic ovary syndrome.
• Congenital adrenal hyperplasia.
• Steroid-producing tumours.
• Precocious puberty/virilization.
• Hirsutism.
Pathophysiology
Urine steroid profile useful in:
• Mineralocortoid hypertension (dK).
• Polycystic ovary syndrome.
• Congenital adrenal hyperplasia.
• Cushing’s Syndrome.
• Androgen resistance.
Further reading
Taylor NF (2006). Urinary steroid profiling. Method Mol Biol 324, 159–75.
PHYSIOLOGY 231
232 CHAPTER 3 Adrenal
Imaging
Computed tomography (CT) scanning
CT is the most widely used modality for imaging the adrenal glands. It is
able to detect masses >5mm in diameter. It can be useful in differentiating
between different adrenal pathologies, in particular by identifying benign
adrenal tumours with high fat content (adrenocortical adenoma, adreno-
myelolipoma), as indicated by pre-contrast tumour density of less than
10HU (Hounsfield units) (see Fig. 3.3).
Magnetic resonance imaging (MRI)
MRI can also reliably detect adrenal masses >5–10mm in diameter and, in
some circumstances, provides additional information to CT, in particular
by determining signal loss in opposed phase, T2-weighted sequences. Rapid
signal loss is indicative of a benign tumour whereas malignant tumours, and
also phaeochromocytomas, show a delay or lack in signal loss.
Ultrasound (US) imaging
US detects masses >20mm in diameter, but normal adrenal glands are not
usually visible, except in children. Body morphology and bowel gas can
provide technical difficulties.
Normal adrenal
Normal adrenal cortex is assessed by measuring limb thickness and
is considered enlarged at >5mm approximately the thickness of the
diaphragmatic crus nearby.
Adrenal
adenoma Adrenal
glands
Radionucleotide imaging
• 123
Iodine-metaiodobenzylguanidine (MIBG) is a guanethidine analogue,
concentrated in some phaeochromocytomas, paragangliomas,
carcinoid tumours, and neuroblastomas, and is useful diagnostically.
A different isotope 131I-MIBG may be used therapeutically, e.g. in
malignant phaeochromocytomas, when the diagnostic imaging shows
uptake.
• 75Se 6B-selenomethyl-19-norcholesterol. This isotope is concentrated in
functioning steroid synthesizing tissue and has, in the past, been used
to image the adrenal cortex. However, high resolution CT and MRI
have largely replaced it in localizing functional adrenal adenomas.
• 123Iodo-metomidate is an isotope that is derived from etomidate, which
is a known 11β-hydroxylase (CYP11B1) inhibitor. Metomidate binds
CYP11B1, which is specifically expressed in the adrenal gland, and thus
iodo-metomidate SPECT can identify adrenocortical tissue.
Positron emission tomography (PET)
PET can be useful in locating tumours and metastases. Radiopharma-
ceuticals for specific endocrine tumours are being developed, e.g.
11
C-metahydroxyephedrine for phaeochromocytoma—combined with CT
(PET-CT), it may offer particular value in localizing occult neuroendocrine
tumours. 11C-metomidate PET-CT has recently been shown to be of value
for the lateralization of aldosterone-producing adrenal masses.
Venous sampling
Adrenal vein sampling (AVS) (b p. 244) can be useful to lateralize an
adenoma or to differentiate an adenoma from bilateral hyperplasia. It
is technically difficult—particularly, catheterizing the right adrenal vein
because of its drainage into the IVC. There is no widespread agreement
on the exact protocol, e.g. consecutive or concurrent adrenal vein cath-
erization, with and without concurrent or preceding ACTH stimulation.
Usually, samples are drawn from the vena cava, superior and inferior to
the renal veins, and separately from both adrenal veins, with subsequent
determination of plasma aldosterone and serum cortisol levels (correct
localization of the catheter in the adrenal vein is only confirmed when
cortisol >3-fold of vena cava cortisol). AVS is of particular value in lateral-
izing small aldosterone-producing adenomas that cannot easily be visual-
ized on CT or MRI.
Further reading
Boland GW (2011). Adrenal imaging: from Addison to algorithms. Radiol Clin North Am 49, 511–28.
Guest P (2011). Imaging of the adrenal gland. In: Wass JAH, Stewart PM (eds.) Oxford textbook of
endocrinology, pp. 763–73. Oxford University Press, Oxford.
Hahner S, Sundin A (2011). Metomidate-based imaging of adrenal masses. Horm Cancer 2, 348–53.
Pacak K, Eisenhofer G, Goldstein DS (2004). Functional imaging of endocrine tumors: role of
positron emission tomography. Endocr Rev 25, 568–80.
234 CHAPTER 3 Adrenal
Primary aldosteronism
Epidemiology
Primary hyperaldosteronism is present in around 10% of hypertensive
patients. It is the most prevalent form of secondary hypertension. The
most common cause is bilateral adrenal hyperplasia (see Table 3.2).
Pathophysiology
Aldosterone causes renal sodium retention and potassium loss. This results
in expansion of body sodium content, leading to suppression of renal renin
synthesis. The direct action of aldosterone on the distal nephron causes
sodium retention and loss of hydrogen and potassium ions, resulting in a
hypokalaemic alkalosis, although serum potassium may not be significantly
reduced and may be normal in up to 50% of cases.
Aldosterone has pathophysiological effects on a range of other tis-
sues, causing cardiac fibrosis, vascular endothelial dysfunction, and
nephrosclerosis.
Clinical features
• Moderately severe hypertension, which is often resistant to
conventional therapy. There may be disproportionate left ventricular
hypertrophy.
• Hypokalaemia is usually asymptomatic. Occasionally, patients may
present with tetany, myopathy, polyuria, and nocturia (hypokalaemic
nephrogenic diabetes insipidus) due to severe hypokalaemia.
• There may be an association with osteoporosis.
PRIMARY ALDOSTERONISM 237
Conn’s syndrome—
aldosterone-producing adenoma
Very high levels of the enzyme aldosterone synthase are expressed in
tumour tissue. Recently, inactivating mutations in the potassium chan-
nel KCJN5 have been identified as the cause of disease in approximately
40% of aldosterone-producing adrenal adenomas. Very rarely, Conn’s
adenomas may be part of the MEN-1 syndrome.
BILATERAL ADRENAL HYPERPLASIA 239
Glucocorticoid-remediable
aldosteronism (GRA)
This is a rare autosomal dominantly inherited condition due to the pres-
ence of a chimeric gene (8q22) containing the 5′ sequence, which deter-
mines regulation of the 11B-hydroxylase gene (CYP11B1) coding for the
enzyme catalysing the last step in cortisol synthesis, and the 3′ sequence
from the aldosterone synthase gene (CYP11B2) coding for the enzyme
catalysing the last step in aldosterone synthesis. This results in the expres-
sion of aldosterone synthase in the zona fasciculata as well as the zona
glomerulosa, and aldosterone secretion comes under ACTH control.
Glucocorticoids are the treatment of choice and lead to suppression of
ACTH and suppression of aldosterone production.
• Early hypertension and family history.
• Hybrid steroids (18OHcortisol and 18oxocortisol) elevated.
ALDOSTERONE-PRODUCING CARCINOMA 241
Aldosterone-producing carcinoma
Rare and usually associated with excessive secretion of other corticoster-
oids (cortisol, androgen, oestrogen). Hypokalaemia may be profound and
aldosterone levels very high. A tumour larger than 2.5cm associated with
aldosterone excess has to be treated as suspicious.
242 CHAPTER 3 Adrenal
Screening
Indications
• Patients resistant to conventional antihypertensive medication (i.e. not
controlled on three agents).
• Hypertension associated with hypokalaemia (potassium <3.7mmol/L,
irrespective of thiazide use). NB: d K only present in 40%.
• Hypertension developing before age of 40 years.
• Adrenal incidentaloma.
Method
• Give oral supplements of potassium to control hypokalaemia;
screening and confirmation should be carried out in the
normokalaemic situation.
• There is no need to stop all concomitant antihypertensive medication
for the first screening by paired plasma aldosterone and plasma
renin measurements; the only medication that must be stopped are
mineralocorticoid receptor antagonists (spironolactone, eplerenone)
that must be stopped 4 weeks prior to diagnostic tests.
• Measure aldosterone:renin ratio.
• A high ratio is suggestive of p hyperaldosteronism (aldosterone
(pmol/L)/plasma renin activity (ng/mL/h) >750 or aldosterone
(ng/dL)/plasma renin activity (ng/mL/h) >30–50). Note that newer
assays that measure renin mass (rather than activity) will require
the development of different cut-offs.
• Test results should be interpreted, having the effects of
antihypertensive drugs in mind (see Table 3.3); B-blockers can cause
false +ves while ACE/AT1R blockers can cause false –ves in borderline
cases. In doubt, the test can be repeated off these drugs; BP can be
controlled using doxazosin or calcium antagonists. False –ve results
can also occur in patients with chronic renal failure due to upregulated
plasma renin.
Confirmation of diagnosis
Confirmation of autonomous aldosterone production is made by demon-
strating failure to suppress aldosterone in face of sodium/volume loading.
This can be achieved by a number of mechanisms after optimizing test
conditions as described.
• Test of choice—saline infusion test:
• Administer 2L normal saline over 4h.
• Measure plasma aldosterone at 0, 2, 3, and 4h.
• Aldosterone fails to suppress to <140pmol/L (140–280 equivocal)
in 80–90% of p aldosteronism.
• Most used test; caution in patients with fluid overload and/or
evidence of heart failure.
• Fludrocortisone suppression test:
• Give fludrocortisone 100 micrograms 6-hourly for 4 days.
• Measure plasma aldosterone basally and on last day.
• Aldosterone fails to suppress in p aldosteronism.
• Caveat: difficult to execute in hypokalaemic, hypertensive patients.
SCREENING 243
Treatment
Surgery
• Laparascopic adrenalectomy is the treatment of choice for
aldosterone-secreting adenomas and is associated with lower
morbidity than open adrenalectomy.
• Surgery is not indicated in patients with idiopathic hyperaldosteronism,
as even bilateral adrenalectomy may not cure the hypertension.
• Presurgical spironolactone treatment may be used to correct
potassium stores before surgery.
• The BP response to treatment with spironolactone (50–400mg/day)
before surgery can be used to predict the response to surgery of
patients with adenomas.
• Hypertension is cured in about 70%.
• If it persists (more likely in those with long-standing hypertension and
increased age), it is more amenable to medical treatment.
• Overall, 50% become normotensive in 1 month and 70% within 1 year.
• Adrenal carcinoma. Open surgery and post-operative adrenolytic
therapy with mitotane is usually required, but the prognosis is usually
poor (b see Treatment, p. 252).
Medical treatment
Medical therapy remains an option for patients with bilateral disease and
those with a solitary adrenal adenoma who are unlikely to be cured by
surgery, who are unfit for operation, or who express a preference for
medical management.
• The mineralocorticoid receptor antagonist spironolactone
(50–400mg/day; once daily administration) has been used successfully
for many years to treat the hypertension and hypokalaemia associated
with bilateral adrenal hyperplasia and idiopathic hyperaldosteronism
profile.
• There may be a delay in response of hypertension of 4–8 weeks.
However, combination with other antihypertensive agents (ACEI and
calcium channel blockers) is usually required.
• Spironolactone also interacts with the androgen and the progesterone
receptor, which contributes to its side effect. Side effects are
common—particularly, gynaecomastia and impotence in ♂, menstrual
irregularities in ♀, and GI effects.
• Eplerenone (50–200mg/day; twice daily administration) is a
mineralocorticoid receptor antagonist without antiandrogen effects
and hence greater selectivity and less side effects than spironolactone.
• Alternative drugs include the potassium-sparing diuretics amiloride
and triamterene. Amiloride may need to be given in high dose (up to
40mg/day) in p aldosteronism, and monitoring of serum potassium is
essential. Calcium channel antagonists may also be helpful.
• Glucocorticoid-remediable aldosteronism can be treated with
low-dose dexamethasone (0.25–0.5mg on going to bed). If needed,
spironolactone and/or amiloride can be added.
TREATMENT 247
Further reading
Allolio B, Hahner S, Weismann D, et al. (2004). Management of adrenocortical carcinoma. Clin
Endocrinol 60, 273–8.
Espiner EA, Ross DG, Yandle TG, et al. (2003). Predicting surgically remedial primary aldosteron-
ism: role of adrenal scanning, posture testing, and adrenal vein sampling J Clin Endocrinol Metab
88, 3637–44.
Funder JW, et al. (2008). Case detection, diagnosis, and treatment of patients with primary aldo-
steronism: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 93, 3266–81.
Ganguly A (1998). Primary aldosteronism. N Engl J Med 339, 1828–33.
Mulatero P (2002). Drug effects on aldosterone/plasma renin activity ratio in primary aldosteronism.
Hypertension 40, 897–902.
Young WF (2007). Primary aldosteronism—renaissance of a syndrome. Clin Endocrinol 66, 607–18.
248 CHAPTER 3 Adrenal
Liquorice ingestion
• Liquorice contains glycyrrhetinic acid which is used as a sweetener.
• It inhibits the action of 11B-HSD2, which allows circulating cortisol
to get increased access to the mineralocorticoid receptor causing
hypertension.
• Liquorice can be found in sweets, chewing tobacco, cough mixtures,
and some herbal medicines.
Ectopic ACTH syndrome
(b see p. 164.)
In the syndrome of ectopic ACTH and generally in very severe Cushing’s
cases, the 11B-HSD2 protection is overcome because of high cortisol
secretion rates, which saturate the enzyme, leading to impaired conver-
sion of cortisol to cortisone. Therefore, cortisol has greater access to the
mineralocorticoid receptor in the kidney, which results in hypokalaemia
and hypertension.
Deoxycorticosterone (DOC) excess
Two forms of congenital adrenal hyperplasia (b see p. 320) are asso-
ciated with excess production of deoxycorticosterone (DOC), as they
result in inhibition of enzymes downstream of DOC and thus lead to its
accumulation. DOC acts as an agonist at the mineralocorticoid receptor,
resulting in hypertension with suppression of renin.
• 17α-hydroxylase (CYP17A1) deficiency.
• 11B-hydroxylase (CYP11B1) deficiency.
Glucocorticoid replacement to inhibit ACTH is effective treatment for
these conditions.
Adrenal tumours, in particular adrenocortical carcinomas, rarely secrete
excessive amounts of deoxycorticosterone. This may be concomitant
with excessive aldosterone production, but occasionally it may occur in
an isolated fashion.
250 CHAPTER 3 Adrenal
Adrenal surgery
Adrenalectomy
Open adrenalectomy may still be necessary for large and complex pathol-
ogy. However, laparoscopic adrenalectomy (first performed in 1992) has
become the procedure of choice for removal of most adrenal tumours.
Retrospective comparisons with open approaches suggest reduced hospi-
tal stay and analgesic requirements and lower post-operative morbidity.
This may be improved even further by the recently introduced retroperi-
toneoscopic approach. However, there are few long-term outcome data
on this technique. It is most useful in the management of small (<6cm)
benign adenomas. Laparoscopic bilateral adrenalectomy, although techni-
cally demanding, offers a useful approach to patients with macronodular
hyperplasia and in selected patients with ACTH-dependent Cushing’s syn-
drome where alternative therapeutic options are not appropriate.
Preoperative preparation of patients
• Cushing’s—metyrapone or ketoconazole (b see p. 170).
• Phaeochromocytoma—α- and B-blockade (b see p. 294).
Perioperative management
See Box 3.2.
ADRENAL SURGERY 257
Further reading
Dudley NE, Harrison BJ (1999). Comparison of open posterior versus transperitoneal laparoscopic
adrenalectomy. Br J Surg 86, 656–60.
McCallum R, Connell JMC (2001). Laparoscopic adrenalectomy. Clin Endocrinol 55, 435–6.
Wells SA, Merke DP, Cutler GB, et al. (1998). The role of laparoscopic surgery in adrenal disease.
J Clin Endocrinol Metab 83, 3041–9.
258 CHAPTER 3 Adrenal
Liddle’s syndrome
A rare autosomal dominant (AD) condition with variable penetrance that
is caused by mutations in the gene encoding for the epithelial sodium
channel (ENaC), usually by mutations in the gene encoding the B or G sub-
unit of ENaC, which is highly selective and located in the distal nephron.
This leads to constitutive activation of sodium transport, independent of
circulating mineralocorticoids; consequently, s activation of the sodium/
potassium exchange occurs.
Features
Hypokalaemia and hypertension.
Investigation
• Hypokalaemic alkalosis.
• Suppressed renin and aldosterone levels.
• See Table 3.5.
Treatment
Hypertension responds to amiloride (doses up to 40mg/day) but not
spironolactone because amiloride acts on the sodium channel directly
whereas spironolactone acts on the mineralocorticoid receptor.
BARTTER’S SYNDROME 259
Bartter’s syndrome
Cause
Loss of function of the bumetanide-sensitive Na–K–2Cl co-transporter
in the thick ascending limb of the loop of Henle. Mutations in three
genes have been reported to account for the phenotype—these encode
regulatory ion channels (NKCC2; ROMK; CLCNKB). Inactivation of the
co-transporter leads to salt wasting, activation of the renin–angiotensin
system, and i aldosterone which leads to i sodium reabsorption at the
distal nephron and causes hypokalaemic alkalosis. Reabsorption of calcium
also occurs in the thick ascending loop, and thus inactivation leads to
hypercalciuria. The lack of associated hypertension is thought to be due to
i prostaglandin production from the renal medullary interstitial tissue in
response to hypokalaemia.
• Rare (~1/million) autosomal recessive hypokalaemic metabolic
alkalosis associated with salt wasting and normal or reduced BP.
• Usually present at an early age (<5 years).
Features
• Intravascular volume depletion.
• Seizures.
• Tetany.
• Muscle weakness.
There is also an antenatal variant which is a life-threatening disorder of
renal tubular hypokalaemic alkalosis and hypercalciuria.
Investigations
• Hypokalaemic alkalosis.
• i PRA and aldosterone.
• Hypercalciuria.
Treatment
Potassium replacement. Potassium-sparing diuretics may be helpful. However,
they are usually inadequate in correcting hypokalaemia. Prostaglandin syn-
thase inhibitors (NSAIDs), e.g. indometacin 2–5mg/kg per day or ibuprofen,
may be required.
260 CHAPTER 3 Adrenal
Gitelman’s syndrome
Cause
• Loss of function in the thiazide-sensitive Na–Cl transporter of the
distal convoluted tubule (DCT) due to mutations in the SCL12A3 gene
that encodes it (located on chromosome 16q13). This leads to salt
wasting, hypovolaemia, and metabolic alkalosis.
• Hypovolaemia leads to activation of the renin–angiotensin system and
i aldosterone levels.
• Hypokalaemic alkalosis, in conjunction with hypocalciuria and
hypomagnesaemia.
• Present at older ages without overt hypovolaemia (essentially, a less
severe phenotype of Bartter’s syndrome).
Investigations
• Hypokalaemic alkalosis, in association with i renin and aldosterone.
• Hypomagnesaemia.
• Hypocalciuria.
Treatment
• Potassium and magnesium replacement.
• Potassium-sparing diuretics may be required.
• See Table 3.5.
Further reading
Amirlak I, Dawson KP (2000). Bartter’s syndrome. QJM 93, 207–15.
Furuhashi M, Kitamura K, Adachi M, et al. (2005). Liddle’s syndrome caused by a novel mutation
in the proline-rich PY motif of the epithelial sodium channel B-subunit. J Clin Endocrinol Metab
90, 340–4.
Graziani G, Fedeli C, Moroni L et al. (2010) Gitelman syndrome: pathophysiological and clinical
aspects. QJM 103, 741–8.
Nakamura A, Shimizu C, Yoshida M et al. (2010) Problems in diagnosing atypical Gitelman’s syn-
drome presenting with normomagnesaemia. Clin Endocrinol 72, 272–6.
GITELMAN’S SYNDROME 261
262 CHAPTER 3 Adrenal
Mineralocorticoid deficiency
Epidemiology
Rare, apart from the hyporeninaemic hypoaldosteronism associated with
diabetes mellitus.
Causes
Congenital
• Primary adrenal insufficiency due to:
• CAH—certain types, most commonly 21-hydroxylase deficiency, are
associated with MC deficiency; b see p. 320.
• Congenital lipoid adrenal hyperplasia (CLAH) caused by mutations in
the genes encoding steroidogenic acute regulatory protein (StAR),
responsible for rapid import of cholesterol into the mitochondrion,
and the side chain cleavage protein (CYP11A1), responsible for
the conversion of cholesterol to pregnenolone, i.e. the first step of
steroidogenesis.
• Adrenal hypoplasia congenita (AHC) caused by mutations in the
genes encoding the transcription factors SF-1 (NR5A1) and DAX-1
(NR0B1) that play a crucial role in adrenal development.
• Adrenoleukodystrophy affecting 1/20,000 ♂; very long chain fatty
acids (VLCFA) cannot be oxidized in peroxisomes and accumulate
in tissues and the circulation. CNS symptoms may be absent
initially, in particular, in the milder form adrenomyeloneuropathy,
but progressive demyelination can lead to hypertonic tetraparesis,
dementia, epilepsy, coma, or death (in particular, in the early
childhood onset variant adrenoleukodystrophy).
• Rare inherited disorders of aldosterone biosynthesis.
• Pseudohypoaldosteronism—inherited resistance to the action of
aldosterone (b see p. 665). Autosomal dominant and recessive
forms are described. Usually presents in infancy. Treated with sodium
chloride.
Acquired
• All forms of non-congenital primary adrenal insufficiency—b see
p. 266. In secondary adrenal insufficiency, the adrenals are anatomically
intact, and thus regulation of mineralocorticoid secretion by the renin–
angiotensin–aldosterone (RAA) system is intact.
• Drugs—heparin (heparin for >5 days may cause severe hyperkalaemia
due to a toxic effect on the zona glomerulosa); ciclosporin.
• Hyporeninaemic hypoaldosteronism—interference with the renin–
angiotensin system leads to mineralocorticoid deficiency and
hyperkalaemic acidosis (type IV renal tubular acidosis), e.g. diabetic
nephropathy. Treatment is fludrocortisone and potassium restriction.
ACEI may produce a similar biochemical picture, but here the PRA will
be elevated, as there is no angiotensin II feedback on renin.
Treatment
Fludrocortisone.
MINERALOCORTICOID DEFICIENCY 263
264 CHAPTER 3 Adrenal
Adrenal insufficiency
Definition
Adrenal insufficiency is defined by the lack of cortisol, i.e. glucocor-
ticoid deficiency, and may be due to destruction of the adrenal cortex
(p, Addison’s disease and congenital adrenal hyperplasia (CAH); b see
p. 266) or due to disordered pituitary and hypothalamic function (s).
Epidemiology
• Permanent adrenal insufficiency is found in 5 in 10,000 population.
• The most frequent cause is hypothalamic–pituitary damage, which is
the cause of AI in 60% of affected patients.
• The remaining 40% of cases are due to primary failure of the adrenal
to synthesize cortisol, with almost equal prevalence of Addison’s
disease (mostly of autoimmune origin, prevalence 0.9–1.4 in 10,000)
and congenital adrenal hyperplasia (0.7–1.0 in 10,000).
• s adrenal insufficiency due to suppression of pituitary–hypothalamic
function by exogenously administered, supraphysiological
glucocorticoid doses for treatment of, for example, COPD or
rheumatoid arthritis, is much more common (50–200 in 10,000
population). However, adrenal function in these patients can recover,
following tapering and cessation of exogenous glucocorticoid
administration.
Causes of secondary adrenal insufficiency
Lesions of the hypothalamus and/or pituitary gland
• Tumours—pituitary tumour, metastases, craniopharyngioma.
• Infection—tuberculosis.
• Inflammation—sarcoidosis, histiocytosis X, haemochromatosis,
lymphocytic hypophysitis.
• Iatrogenic—surgery, radiotherapy.
• Other—isolated ACTH deficiency, trauma.
Suppression of the hypothalamo–pituitary–adrenal axis
• Glucocorticoid administration.
• Cushing’s disease (after pituitary tumour removal).
Features of secondary adrenal insufficiency
As p (b see Clinical features, p. 123), except:
• Absence of pigmentation—skin is pale.
• Absence of mineralocorticoid deficiency.
• Associated features of underlying cause, e.g. visual field defects if
pituitary tumour.
• Other endocrine deficiencies may manifest due to pituitary failure
(b see p. 122).
• Acute onset may occur due to pituitary apoplexy.
ADRENAL INSUFFICIENCY 265
Addison’s disease
Causes of primary adrenal insufficiency
• Autoimmune—commonest cause in the developed world
(approximately 70% cases).
• Autoimmune polyglandular deficiency—type 1 or 2 (b see p. 270).
• Malignancy:
• Metastatic (lung, breast, kidney—adrenal metastases found in
~50% of patients, but symptomatic adrenal insufficiency much less
common and only observed with bilateral metastases).
• Lymphoma (primary adrenal lymphoma, AI only if bilateral).
• Infiltration:
• Amyloid.
• Sarcoidosis.
• Haemochromatosis.
• Infection:
• Tuberculosis (medulla more frequently destroyed than cortex).
• Fungal, e.g. histoplasmosis, cryptococcosis.
• Opportunistic infections in, for example AIDS—CMV,
Mycobacterium intracellulare, cryptococcus (up to 5% patients with
AIDS develop p adrenal insufficiency in the late stages).
• Vascular haemorrhage:
• Anticoagulants.
• Waterhouse–Friderichsen syndrome in meningococcal septicaemia.
• Infarction—e.g. s to thrombosis in antiphospholipid syndrome.
• Adrenoleukodystrophy:
• Inherited disorder caused by mutations in the X-linked ALD gene
that encodes for the peroxisomal transporter protein ABCD1.
• Diagnosed by measuring very long chain fatty acids.
• Presents in childhood and adolescence.
• Progresses in 50% to quadriparesis and dementia, in
association with adrenal failure (= cerebral ALS); milder variant
adrenomyeloneuropathy (AMN) causes spinal neurology only, and
AI may precede manifestation of neurological symptoms; in 10–20%,
AI is the sole manifestation of disease.
• Congenital adrenal hyperplasia—b see p. 320.
• Adrenal hypoplasia congenita—very rare familial failure of adrenal
cortical development due to mutations/deletion of the NR0B1
(DAX-1) or the NR5A1 (SF-1) genes.
• Congenital lipoid adrenal hyperplasia—very rare familial failure of
adrenal steroidogenesis due to mutations in the genes encoding
the steroidogenic acute regulatory protein (StAR; responsible for
mitochondrial import of cholesterol) or the side chain cleavage
enzyme CYP11A1 (responsible for conversion of cholesterol to
pregnenolone, i.e. the first step of steroidogenesis).
ADDISON’S DISEASE 267
Autoimmune adrenalitis
• Mediated by humoral and cell-mediated immune mechanisms.
Autoimmune insufficiency associated with polyglandular autoimmune
syndrome is more common in ♀ (70%).
• Adrenal cortex antibodies are present in the majority of patients at
diagnosis, and although titres decline and eventually disappear, they are
still found in approximately 70% of patients 10 years later. Up to 20%
patients/year with +ve adrenal antibodies develop adrenal insufficiency.
Antibodies to 21-hydroxylase are commonly found, although the exact
nature of other antibodies that block the effect of ACTH, for example,
is yet to be elucidated.
• Antiadrenal antibodies are found in <2% of patients with other
autoimmune endocrine disease (Hashimoto’s thyroiditis, diabetes
mellitus, autoimmune hypothyroidism, hypoparathyroidism, pernicious
anaemia). In addition, antibodies to other endocrine glands are
commonly found in patients with autoimmune adrenal insufficiency
(thyroid microsomal in 50%, gastric parietal cell, parathyroid, and ovary
and testis). However, the presence of antibodies does not predict
subsequent manifestation of organ-specific autoimmunity.
• Polyglandular autoimmune conditions (b see Autoimmune
polyglandular syndrome (APS) type 1, p. 270; APS type 2, p. 271).
The presence of 17-hydroxylase antibodies, in association with
21-hydroxylase antibodies, is a good marker of patients at risk of
developing premature ovarian failure in association with p adrenal
failure.
• Patients with type 1 diabetes mellitus and autoimmune thyroid disease
only rarely develop autoimmune adrenal insufficiency. Approximately
60% of patients with Addison’s disease have other autoimmune or
endocrine disorders.
Clinical features
Chronic
• Anorexia and weight loss (>90%).
• Tiredness.
• Weakness—generalized, no particular muscle groups.
• Pigmentation—generalized but most common in skin areas exposed
to friction or pressure (elbows and knees, and under bras and belts),
mucosae, and scars acquired after onset of adrenal insufficiency. Look
at palmar creases in Caucasians.
• Dizziness and postural hypotension.
• GI symptoms—nausea and vomiting, abdominal pain, diarrhoea.
• Arthralgia and myalgia.
• Symptomatic hypoglycaemia—rare in adults.
• d axillary and pubic hair and reduced libido in ♀.
• Pyrexia of unknown origin—rarely.
Associated conditions
• Vitiligo.
• Features of other autoimmune endocrinopathies.
AUTOIMMUNE ADRENALITIS 269
Laboratory investigations
• Hyponatraemia.
• Hyperkalaemia.
• Elevated urea.
• Anaemia (normocytic normochromic).
• Elevated ESR.
• Eosinophilia.
• Mild hypercalcaemia—d absorption, d renal absorption of calcium.
270 CHAPTER 3 Adrenal
Autoimmune polyglandular
syndrome (APS) type 1
• Also known as autoimmune polyendocrinopathy, candidiasis, and
ectodermal dystrophy (APECED).
• Autosomal recessive with childhood onset.
• Chronic mucocutaneous candidiasis.
• Hypoparathyroidism (90%), p adrenal insufficiency (60%).
• p gonadal failure (41%)—usually after Addison’s diagnosis.
• p hypothyroidism.
• Rarely hypopituitarism, diabetes insipidus, type 1 diabetes mellitus.
• Associated chronic active hepatitis (20%), malabsorption (15%),
alopecia (40%), pernicious anaemia, vitiligo.
• Mutations in the AIRE (autoimmune regulator) gene located on
chromosome 21p22.3.
APS TYPE 2 271
APS type 2
• Polygenic inheritance, association with HLADR3 and CTL4 regions,
mixed penetrance.
• Adult onset.
• Adrenal insufficiency (100%).
• p autoimmune thyroid disease (70%), mostly hypothyroidism but also
hyperthyroidism.
• Type 1 diabetes mellitus (5–20%)—often before Addison’s diagnosis.
• p gonadal failure in affected women (5–20%).
• Rarely diabetes insipidus (<0.1%).
• Associated vitiligo, myasthenia gravis, alopecia, pernicious anaemia,
immune thrombocytopenic purpura.
Eponymous syndromes
• Schmidt’s syndrome:
• Addison’s disease, and
• Autoimmune hypothyroidism.
• Carpenter syndrome:
• Addison’s disease, and
• Autoimmune hypothyroidism, and/or
• Type 1 diabetes mellitus.
272 CHAPTER 3 Adrenal
Intercurrent illness
• Cortisol requirements increase during severe illness or surgery.
• For moderate elective procedures or investigations, e.g. endoscopy
or angiography, patients should receive a single dose of 100mg
hydrocortisone before the procedure.
• For major surgery, patients should receive 100mg intravenously or
intramuscularly with the premedication and receive:
• 50–100mg IM hydrocortisone 6-hourly for the first 3 days, or
• 100–150mg per 24h IV in 5% glucose before reverting rapidly to a
maintenance dose.
• To cover severe illness, e.g. pneumonia, patients should receive
50–100mg IM hydrocortisone 6-hourly until resolution of the illness.
• See Box 3.4 for pregnancy.
Drug interactions
• Rifampicin:
• Increases the clearance of cortisol.
• Double usual dose of hydrocortisone.
• Mitotane:
• Increases cortisol-binding globulin and induces CYP3A4, resulting in
rapid inactivation of hydrocortisone.
• Double usual dose of hydrocortisone, e.g. 20–10–10mg.
• Topiramate—also induces CYP3A4 and accelerates clearance of
glucocorticoids.
Further reading
Arlt W (2009). The approach to the adult with newly diagnosed adrenal insufficiency. J Clin
Endocrinol Metab 94, 1059–67.
Arlt W, Allolio B (2003). Adrenal insufficiency. Lancet 361, 1881–93.
Bornstein SR (2009). Predisposing factors for adrenal insufficiency. N Engl J Med 360, 2328–39.
Lebbe M, Arlt W (2013). What is the best diagnostic and therapeutic management strategy for an
Addison patient during pregnancy? Clin Endocrinol (Oxf) 78, 497–502.
Mitchell AL, Pearce SH (2012). Autoimmune Addison disease: pathophysiology and genetic
complexity. Nat Rev Endocrinol 8, 306–16.
Wass JAH, Arlt W (2012). How to avoid precipitating an acute adrenal crisis. BMJ 345, 6333.
TREATMENT OF PRIMARY ADRENAL INSUFFICIENCY 279
280 CHAPTER 3 Adrenal
Long-term glucocorticoid
administration
Both exogenous glucocorticoid administration and endogenous excess
glucocorticoids (Cushing’s syndrome) lead to a –ve feedback effect on
the hypothalamo–pituitary axis (HPA), leading to suppression of both
CRH and ACTH secretion and atrophy of the zonae fasciculata and
reticularis of the adrenal cortex. Administration (or endogenous over-
production) of >50mg hydrocortisone equivalent per day will result in
downregulation of mineralocorticoid production. However, the renin–
angiotensin–aldosterone axis continues to function when the glucocorti-
coid excess ceases while the HPA axis will suffer from lasting suppression,
with the time of recovery dependent on the duration of the preceding
period of hypercortisolism. See Box 3.6 for steroid equivalents.
Short-term steroids
• Any patient who has received glucocorticoid treatment for <3 weeks
is unlikely to have clinically significant adrenal suppression, and if the
medical condition allows it, glucocorticoid treatment can be stopped
acutely. A major stress within a week of stopping steroids should,
however, be covered with glucocorticoids.
• Exceptions to this are patients who have other possible reasons for
adrenocortical insufficiency, who have received >40mg prednisolone
(or equivalent), where a short course has been prescribed within
1 year of cessation of long-term therapy, or evening doses (i HPA axis
suppression).
Steroid cover
While receiving glucocorticoid treatment and within 1 year of steroid
withdrawal, patients should receive standard steroid supplementation at
times of stress, e.g. major trauma, surgery, and infection (b see p. 277).
Long-term steroids
• When patients are receiving supraphysiological doses (>5mg
prednisolone or equivalent) of glucocorticoid, dose reduction depends
on the activity of the underlying disease requiring glucocorticoid
treatment.
• If the disease has resolved, the dose can be rapidly reduced to 5mg
prednisolone by a reduction of 2.5mg every 3–5 days.
• Once the patient is established on 5mg prednisolone, consider
changing to hydrocortisone (20mg in the morning), as this has a
shorter half-life and will, therefore, lead to less prolonged suppression
of ACTH.
• Daily hydrocortisone dose should be reduced by 2.5mg every
1–2 weeks, or as tolerated, until a dose of 10mg is reached.
After 2–3 months, a short Synacthen® test should be performed.
Once a short Synacthen® test demonstrates a normal response,
hydrocortisone replacement can be stopped. Supplemental steroids
during intercurrent illness are not required.
LONG-TERM GLUCOCORTICOID ADMINISTRATION 281
Cushing’s syndrome
Patients with Cushing’s syndrome on metyrapone or with recently treated
disease, whatever the cause, may also have HPA axis suppression and may,
therefore, need steroid replacement at times of stress.
Adrenal incidentalomas
Definition and epidemiology
• An adrenal incidentaloma is an adrenal mass that is discovered
incidentally upon imaging (e.g. CT chest or abdomen) carried out for
reasons other than a suspected adrenal pathology.
• The incidental detection of an adrenal mass is becoming more
common, as i numbers of imaging procedures are performed and with
technological improvements in imaging.
• Autopsy studies suggest incidence prevalence of adrenal masses of
1–6% in the general population.
• Imaging studies suggest that adrenal masses are present 2–3% in the
general population. Incidence increases with ageing, and 8–10% of
70-year olds harbour an adrenal mass.
Importance
It is important to determine whether the incidentally discovered adrenal
mass is:
• Malignant.
• Functioning and associated with excess hormonal secretion.
Differential diagnosis of an incidentally detected
adrenal nodule
• Cortisol-secreting adrenal adenoma causing Cushing’s syndrome or
subclinical Cushing’s syndrome (5%).
• Mineralocorticoid-secreting adrenal adenoma.
• CAH.
• Adrenocortical carcinoma (5–12%).
• Metastasis (2–10%; most prevalent breast, lung, kidney).
• Phaeochromocytoma (10–15%).
• Adrenal cysts (5%).
• Adrenal myelolipoma (5–10%).
• Haematoma.
• Ganglioneuroma (4%).
Investigations
• Clinical assessment for symptoms and signs of excess hormone
secretion and signs of extra-adrenal carcinoma.
• Urinary free cortisol and overnight dexamethasone suppression test.
• Plasma free metanephrines (most sensitive and specific screening test;
alternatively, urinary metanephrine—however, more cumbersome and
less specific) (b see p. 288).
• Aldosterone/renin ratio and serum potassium.
• A homogeneous mass with a low attenuation value prior to contrast
administration (<10HU) on CT scan is likely to be a benign adenoma
(low density = high fat content = benign) whereas a mass with high
density (>20HU) has to be considered suspicious (differential diagnosis
phaeochromocytoma/adrenocortical carcinoma/metastasis but also
lipid-poor adenoma).
ADRENAL INCIDENTALOMAS 283
Phaeochromocytomas and
paragangliomas
Definition
• Phaeochromocytomas (aPCA) are chromaffin tumours arising from the
adrenal medulla and secreting catecholamines.
• Paragangliomas are tumours arising from extra-adrenal sympathetic or
parasympathetic nervous tissue.
• Sympathetic paraganglia occur as follows: in prevertebral, paravertebral,
thoracoabdominal, in the pelvis area and close to reproductive organs,
prostate, bladder, liver, and the organ of Zuckerkandl (at the bifurcation
of the aorta). Tumours arising from this sympathetic tissue are termed
extra-adrenal functional paraganglioma (eFPGL).
Parasympathetic paraganglia are located close to major arteries and
nerves, e.g. carotid body, glomus jugulare, vagal, tympanic, pulmonary,
and aorta. Tumours arising from the parasympathetic nervous system are
referred to as head and neck paraganglioma (HNPGL), and only a minority
of those shows endocrine activity (~25%).
Incidence
Rare tumours, accounting for <0.1% of causes of hypertension. However,
it is a very important diagnosis due to:
• The development of potentially fatal hypertensive crises.
• The reversibility of all its manifestations after surgical removal of the
tumour.
• The lack of long-term efficacy of medical treatment.
• The appreciable incidence of malignancy.
• The implications of the identification of an underlying genetic cause
(>25% of cases, e.g. RET mutations and co-incident medullary
carcinoma, VHL mutations and co-incident angiomas, or SDHB
mutations and their potential for recurrent and malignant tumours).
Epidemiology
• Equal sex distribution, and most commonly present in the third and
fourth decades. Up to 50% may be diagnosed post-mortem.
• Tumours may be bilateral, particularly where part of an inherited
syndrome (see Table 3.6).
• 10% of aPCA/eFPGL occur in patients with a family history of
phaeochromocytoma, and currently 25% of patients with apparently
sporadic phaeochromocytoma are found to harbour a disease-causing
mutation upon genetic analysis.
Multiple tumours, extra-adrenal location, or evidence of malignancy are
indicators of a high likelihood of an underlying genetic cause. See Box 3.7
for who should be screened.
PHAEOCHROMOCYTOMAS AND PARAGANGLIOMAS 285
Pathophysiology
Sporadic tumours are usually unilateral and <10cm in diameter. Tumours
associated with familial syndromes are more likely to be bilateral and asso-
ciated with pre-existing medullary hyperplasia.
Malignancy
• Approximately 15–20% are malignant, and these are characterized by
local invasion or distant metastasis rather than capsular invasion.
• Differentiating benign and malignant tumours is difficult and mainly
based on the presence of metastases, although chromosomal ploidy
may be useful.
• Paragangliomas are more likely to be malignant and to recur.
• Typical sites for metastases are retroperitoneum, lymph nodes, bone,
liver, and mediastinum.
Secretory products
• Catecholamine secretion is usually adrenaline or noradrenaline and
may be constant or episodic.
• Phenylethanolamine-N-methyltransferase (PNMT) is necessary for
methylation of noradrenaline to adrenaline and is cortisol-dependent.
• Paragangliomas (exception—organ of Zuckerkandl) secrete
noradrenaline only, as they lack PNMT.
• Small adrenal tumours tend to produce more adrenaline whereas
larger adrenal tumours produce more noradrenaline, as a proportion
of their blood supply is direct, rather than corticomedullary, and
therefore, lower in cortisol concentrations.
• Pure dopamine secretion is rare and may be associated with
hypotension. These tumours are more likely to be malignant.
• Other non-catecholamine secretory products may also be produced,
including VIP, neuropeptide Y, ACTH (associated with Cushing’s
syndrome), PTH, and PTHrP.
PHAEOCHROMOCYTOMAS AND PARAGANGLIOMAS 287
Clinical features
• Sustained or episodic hypertension often resistant to conventional
therapy (BUT: more and more phaeochromocytomas are found
incidentally following CT imaging, thus may not show any clinical signs
and symptoms).
• The presence of palpitation, headaches, or sweating in a
patient with hypertension should raise the diagnostic query of
phaeochromocytoma.
• General:
• Sweating and heat intolerance >80%.
• Pallor or flushing.
• Feeling of apprehension.
• Pyrexia.
• Neurological—headache (throbbing or constant) (65%), paraesthesiae,
visual disturbance, seizures.
• Cardiovascular—palpitations (65%), chest pain, dyspnoea, postural
hypotension.
• GI—abdominal pain, constipation, nausea.
• Skin—livedo reticularis.
• Endocrine—paroxysmal thyroid swelling (noradrenaline-secreting).
Complications
• Cardiovascular—left ventricular failure, dilated cardiomyopathy
(reversible), dysrhythmias.
• Respiratory—pulmonary oedema.
• Metabolic—carbohydrate intolerance, hypercalcaemia.
• Neurological—cerebrovascular, hypertensive encephalopathy.
Factors precipitating a crisis
• Straining.
• Exercise.
• Pressure on abdomen—tumour palpation, bending over.
• Surgery.
• Drugs:
• Anaesthetics.
• Unopposed B-blockade.
• IV contrast agents.
• Opiates.
• Tricyclic antidepressants.
• Phenothiazines.
• Metoclopramide.
• Glucagon.
288 CHAPTER 3 Adrenal
Investigations of phaeochromocytomas
and paragangliomas
Demonstrate catecholamine hypersecretion
24h urine collection is the standard test for screening for a phaeochro-
mocytoma. In a patient with suggestive symptoms, this is usually suffi-
cient to confirm or exclude the diagnosis. False –ves are more common
when patients are asymptomatic and early in the disease. Particular care
is needed in familial cases, incidentalomas, and in those in whom a gen-
eral anaesthetic has precipitated a hypertensive episode. Metadrenalines
(either urine or plasma) offer more specific diagnostic tools than measure-
ment of unmetabolized catecholamines and provide the best biochemical
tests for diagnosing phaeochromocytoma. See Table 3.8 for sensitivity and
specificity of tests.
24h urine collection for catecholamines/metanephrines (see Tables 3.7
and 3.8)
• Urine is collected into bottles containing acid (warn patient).
• Because of the episodic nature of catecholamine secretion, at least
2× 24h collections should be performed. It is useful to perform a
collection while a patient is having symptoms if episodic secretion is
suspected.
• The sensitivity of urinary VMAs is less than free catecholamines or
metadrenalines and also influenced by dietary intake and should not be
used. Urinary metanephrines are of similar sensitivity but of superior
specificity to urinary catecholamines.
• NB Tricyclic antidepressants and labetalol interfere with adrenaline
measurements and should be stopped for 4 days (Box 3.8).
• For marginal elevation, urine collections should be repeated, avoiding
nuts (walnuts), fruit (bananas and pineapple), potatoes, and beans.
Plasma metanephrine measurement
• Plasma metanephrines are the most sensitive test for detection of
catecholamine excess and have only slightly lower specificity than
urinary metanephrines (Table 3.8).
• If plasma metanephrines are borderline, urinary metanephrines may be
used for confirmation.
• Routine measurement requires controlled conditions—supine and
cannulated for 30min; however, false +ves are rare, thus screening
can be carried out in the sitting position without 30min rest, and only
borderline positive cases require repeat after 30min supine rest.
• Plasma catecholamines are elevated by renal failure, caffeine, nicotine,
exercise, and some drugs.
• Catecholamine levels in asymptomatic individuals investigated for
an adrenal incidentaloma or due to a familial condition are often
diagnosed at an earlier stage and may, therefore, have lower
catecholamines.
INVESTIGATIONS OF PHAEOCHROMOCYTOMAS 289
Management
Medical
• It is essential that any patient is fully prepared with α- and B-blockade
before receiving IV contrast or undergoing a procedure, such as
venous sampling or surgery.
• α-blockade must be commenced before B-blockade to avoid
precipitating a hypertensive crisis due to unopposed α-adrenergic
stimulation.
• α-blockade—commence phenoxybenzamine as soon as diagnosis
made. Start at 10mg 2× day by mouth, and increase up to 20mg 4× day
(doxazosin is an accepted alternative).
• B-blockade—use a B-blocker, such as propranolol 20–80mg 8-hourly
by mouth, 48–72h after starting phenoxybenzamine and with evidence
of adequate α-blockade (generally noted by a postural fall in BP).
• Treatment is commenced in hospital. Monitor BP, pulse, and
haematocrit. The goal is a BP of 130/80 or less sitting and 100mg
systolic standing, pulse 60–70 sitting and 70–80 standing. Reversal of
α-mediated vasoconstriction may lead to haemodilution (check Hb
preoperatively).
• To ensure complete blockade before surgery, IV phenoxybenzamine
(1mg/kg over 4h in 100mL 5% glucose) can be administered on the
3 days before surgery. There is less experience with competitive
α-adrenergic blockade, such as prazosin.
• See Table 3.9 for contraindicated drugs.
Surgical (also considered in b see p. 610)
• Surgical resection is curative in the majority of patients, leading to
normotension in at least 75%.
• Mortality from elective surgery is <2%. It is essential that the
anaesthetic and surgical teams have expertise of management of
phaeochromocytomas perioperatively.
• Surgery may be laparoscopic if the tumour is small and apparently
benign. Careful perioperative anaesthetic management is essential,
as tumour handling may lead to major changes in BP and also
occasionally cardiac arrythmias. Phentolamine, nitroprusside, or IV
nicardipine are useful to treat perioperative hypertension, and esmolol
or propranolol for perioperative arrythmias. Hypotension (e.g. after
tumour devascularization) usually responds to volume replacement but
occasionally requires inotropic support.
• Risk factors for haemodynamic instability during surgery include a high
noradrenaline concentration, large tumour size, postural drop after
B-blockade, and a mean arterial pressure >100mmHg.
MANAGEMENT 295
Follow-up
• Cure is assessed by 24h urinary free catecholamine measurement, but
since catecholamines may remain elevated for up to 10 days following
surgery, these should not be performed until 2 weeks post-operatively.
• Lifelong follow-up is essential to detect recurrence of a benign tumour
or metastasis from a malignant tumour, as it is impossible to exclude
malignancy on a histological specimen.
• Chromogranin A is also a useful marker (falsely elevated with proton
pump therapy, steroids, liver and renal failure, essential hypertension,
atrophic gastritis, prostatic carcinoma, and thyrotoxicosis).
Malignancy
• Malignant tumours require long-term α- and B-blockade. The tyrosine
kinase inhibitor α-methylparatyrosine may help control symptoms.
• High-dose 131I-MIBG can be used to treat metastatic disease.
• Chemotherapy, using cyclophosphamide, vincristine, doxorubicin, and
dacarbazine, has been associated with symptomatic improvement.
• Radiotherapy can be useful palliation in patients with bony metastases.
• Novel treatment options for malignant phaeochromocytoma are
urgently needed, and current studies explore VEGF inhibitors on the
basis of the highly vascularized nature of the tumours.
296 CHAPTER 3 Adrenal
Prognosis
• Hypertension may persist in 25% patients who have undergone
successful tumour removal.
• 5-year survival for ‘benign’ tumours is 96%, and the recurrence rate
is <10%.
• 5-year survival for malignant tumours is 44%.
• SHB gene mutation patients are associated with a shorter survival.
Further reading
Astuti D, Latif F, Dallol A, et al. (2001). Gene mutations in the succinate dehydrogenase subunit
SDHB cause susceptibility to familial pheochromocytoma and to familial paraganglioma. Am J
Hum Genet 69, 49–54.
Ayala-Ramirez M, Chougnet CN, Habra MA, et al. (2012). Treatment with sunitinib for patients
with progressive metastatic pheochromocytomas and sympathetic paragangliomas. J Clin
Endocrinol Metab 97, 4040–50.
Eisenhofer G, Lenders JW, Timmers H, et al. (2011). Measurements of plasma methoxytyramine,
normetanephrine and metanephrine as discriminators of different hereditary forms of phaeo-
chromocytoma. Clin Chem 57, 411–20.
Erickson D, Kudva YC, Ebersold MJ, et al. (2001). Benign paragangliomas: clinical presentation and
treatment outcomes in 236 patients. J Clin Endocrinol Metab 86, 5210–16.
Gimm O, Armanios M, Dziema H, et al. (2000). Somatic and occult germ-line mutations in SDHD,
a mitochondrial complex II gene, in nonfamilial phaeochromocytoma. Cancer Res 60, 6822–5.
Ilias I, Yu J, Carrasquillo JA, et al. (2003). Superiority of 6-[18F]-fluorodopamine positron emission
tomography versus [131I]-metaiodobenzylguanidine scintigraphy in the localization of metastatic
pheochromocytoma. J Clin Endocrinol Metab 88, 4083–7.
Jafri M, Maher ER (2012). The genetics of phaeochromocytoma: using clinical features to guide
genetic testing. Eur J Endocrinol 166,151–8.
Lenders JW, Eisenhofer G, Mannelli M, Pacak K (2005). Phaeochromocytoma. Lancet 366, 665–75.
Neumann HP, Bausch B, McWhinney SR, et al. (2002). Germ-line mutations in nonsyndromic
pheochromocytoma. N Engl J Med 346, 1459–66.
Chapter 4 297
Reproductive
endocrinology
Reproductive physiology
Anatomy
• Normal adult ♂ testicular volume 15–30mL.
• Normal adult ♀ ovarian volume 5–10mL.
• Two gonadotrophins LH and FSH address two gonadal cell types, with
feedback from sex steroids and inhibin.
• Three important cells of the gonad:
• Interstitial cells.♂ Leydig cells, ♀ theca cells—which are found
in between the seminiferous tubules and follicles, respectively.
Produce testosterone under LH drive.
• Cells supporting gametogenesis. ♂ Sertoli cells, ♀ granulosa
cells—secrete various hormones, including inhibin and Müllerian
inhibitory factor (AMH) under FSH drive. The former inhibits FSH
secretion from the pituitary gland, and the latter is responsible for
suppressing ♀ sex organ development during sexual differentiation
in utero. In adult ♀, AMH is produced in proportion to germ cell
number and is, therefore, a marker of ovarian ageing.
• Germ cells.♂ continue to make new germ cells throughout adult
life in the basal membrane of tubules. ♀ cease to make new germ
cells after birth and are, therefore, born with all of the ‘eggs’ that
they will ever make.
• Functional units:
• ♂ seminiferous tubules. Make up 90% of testicular volume.
Spermatogenesis occurs here in the presence of high intratesticular
concentrations of testosterone. Made up of germ cells and Sertoli
cells through which spermatogonia mature to be released into the
lumen of the tubule.
• ♀ Graafian follicle. Primordial follicles are recruited in batches,
mature over 2 months, with selection of a dominant follicle with
single central oocyte surrounded by granulosa cells which convert
theca-derived testosterone to oestradiol for ovulation. Follicles
which do not proceed to ovulation become atretic.
See Fig. 4.6 for the sex steroid biosynthesis pathway.
REPRODUCTIVE PHYSIOLOGY 299
Cholesterol
StAR
17α HSD 17, 20 lyase
Pregnenolone 17OH Pregnenolone Dehydroepiandrosterone
3β HSD 17β HSD Aromatase
Progesterone 17OH Progesterone Androstenedione Testosterone Oestradiol
21 OH
Deoxy- 11-Deoxycortisol
corticosterone
11β OH
Corticosterone Cortisol
18-OH Corticosterone
Aldosterone
through the epididymis. This means that events which may affect
spermatogenesis may not be apparent for up to 3 months, and
successful induction of spermatogenesis treatment may take 2 years.
• ♀. From primordial follicle to primary follicle, it takes about 180 days
(a continuous process). It is then another 60 days to form a preantral
follicle which then proceeds to ovulation three menstrual cycles later.
Only the last 2–3 weeks of this process is under gonadotrophin drive,
during which time the follicle grows from 2 to 20mm.
Pituitary cycle
FSH LH
Ovarian cycle
E2
P
Endometrial cycle
M
−12 −8 −4 0 +4 +8 +12
Reproductive physiology
Sex steroid transport
Testosterone daily production rate in ♂ is 5–15mg; 2–4% of total tes-
tosterone circulates as free biologically active hormone. The rest is
bound to proteins, particularly albumin and sex hormone-binding globulin
(SHBG). Several equations are used to estimate free testosterone: free
androgen index ((total testosterone/SHBG) × 100) is of limited value, and
web-based calculators of bioactive testosterone have better validation.
Androstenedione is only about 6% SHBG-bound.
Oestradiol daily production rate in ♀ is 40–400 micrograms; 2–3% free
oestradiol is biologically active; the rest is bound to SHBG.
Sex steroid metabolism
• Testosterone is converted in target tissues to the more potent
androgen DHT in the presence of the enzyme 5A-reductase. There are
multiple 5A-reductase isoenzymes; type 2 is the isoenzyme responsible
for DHT synthesis in the genitalia, genital skin, and hair follicles. It is,
therefore, essential for normal ♂ virilization and sexual development.
• Testosterone may alternatively be converted into oestradiol through
the action of the aromatase enzyme, found in greatest quantities in
testes and adipose tissue.
• Many effects previously attributed to testosterone are now known
to be mediated by oestrogen—especially closure of epiphyses and
maintenance of bone density.
• Testosterone and its metabolites are inactivated in the liver and
excreted in the urine.
• Oestradiol is conjugated in the liver to sulphates and glucuronates and
then extracted in the urine.
Androgen action
• Both testosterone and DHT exert their activity by binding to androgen
receptors, the latter more avidly than testosterone.
• ♂ sexual differentiation during embryogenesis.
• Development and maintenance of ♂s sex characteristics after
puberty.
• Normal ♂ sexual function and behaviour.
• Spermatogenesis.
• Regulation of gonadotrophin secretion.
Oestrogen action
• Oestradiol binds to A (reproductive tissues) and B (bone, brain, heart,
etc.) receptors.
• Development of ♀s sex characteristics.
• Increase fat stores.
• Increase vaginal wall and uterine thickening.
Further reading
De Ronde W, Pols HAP, Van Leeuwen JPTM, et al. (2003). The importance of oestrogens in males.
Clin Endocrinol 58, 529–42.
REPRODUCTIVE PHYSIOLOGY 303
304 CHAPTER 4 Reproductive endocrinology
Hirsutism
Definition
Hirsutism (not a diagnosis in itself) is the presence of excess hair growth in
♀ as a result of i androgen production and i skin sensitivity to androgens.
See Table 4.1 for causes. See Box 4.1 for signs of virilization.
Physiology of hair growth
Before puberty, the body is covered by fine unpigmented hairs or vel-
lus hairs. During adolescence, androgens convert vellus hairs into coarse,
pigmented terminal hairs in androgen-dependent areas. The extent
of terminal hair growth depends on the concentration and duration of
androgen exposure as well as on the sensitivity of the individual hair
follicle. Idiopathic hirsutism refers to those with normal investigations and
presumably greater than average androgen receptor sensitivity.
The reason different body regions respond differently to the same
androgen concentration is unknown but may be related to the number of
androgen receptors in the hair follicle. Genetic factors play an important
role in the individual susceptibility to circulating androgens, as evidenced
by racial differences in hair growth.
Androgen production in women
In ♀, testosterone is secreted primarily by the ovaries and adrenal glands,
although a significant amount is produced by the peripheral conversion of
androstenedione and DHEA. Ovarian androgen production is regulated
by luteinizing hormone, whereas adrenal production is ACTH-dependent.
The predominant androgens produced by the ovaries are testosterone
and androstenedione, and the adrenal glands are the main source of
DHEA. Circulating testosterone is mainly bound to sex hormone-binding
globulin (SHBG), and it is the free testosterone which is biologically active.
Testosterone is converted to dihydrotestosterone in the skin by the
enzyme 5A-reductase. Androstenedione and DHEA are not significantly
protein-bound. See Fig. 4.1.
HIRSUTISM 305
Pituitary
ACTH LH
Adrenals Ovaries
Evaluation of hirsutism
(See Box 4.2 for androgen-independent hair growth.)
Androgen-dependent hirsutism
Normally develops following puberty. Hairs are coarse and pigmented and
typically grow in ♂ pattern. It is often accompanied by other evidence
of androgen excess, such as acne, oily skin and hair, and male pattern
alopecia.
History
• Age and rate of onset of hirsutism. Slowly progressive hirsutism
following puberty suggests a benign cause, whereas rapidly progressive
hirsutism of recent onset requires further immediate investigation to
rule out an androgen-secreting neoplasm.
• Menstrual history. ?oligomenorrhoeic.
• Presence of other evidence of hyperandrogenism, e.g. acne or
bitemporal hair recession.
• Drug history. Some progestins used in oral contraceptive preparations
may be androgenic (e.g. norethisterone).
• Treatments are often based on subjective appearance, so be cautious if
there is a great disparity between subjective and objective assessment.
Consider psychological background.
Physical examination
• Distinguish between androgen-dependent and androgen-independent hair
growth.
• Assess the extent and severity of hirsutism. The Ferriman–Gallwey
score assesses the degree of hair growth in 11 regions of the body.
This provides a semi-objective method of monitoring disease
progression and treatment outcome but is mainly used in research
rather than routine practice.
• Virilization should be looked for only in suspected cases of severe
hyperandrogenism (b see Box 4.1, p. 305).
• Acanthosis nigricans is indicative of insulin resistance and
probable PCOS.
• Rare causes of hyperandrogenism, such as Cushing’s syndrome and
acromegaly, should be ruled out.
Laboratory investigation
Serum testosterone should be measured in all ♀ presenting with hir-
sutism. If this is <5nmol/L, then the risk of a sinister cause for her hirsutism
is low. Further investigations and management of the individual disorders
will be discussed in the following chapters.
Imaging
Pelvic ultrasound may be useful to diagnose PCOS. Idiopathic hirsutism
refers to those with normal ovarian morphology, but this distinction is
not clear-cut, as the level of detection of PCO morphology on US is
operator-dependent.
Further reading
Koulori O, Conway G (2009). Management of hirsutism. BMJ 338, 823–6.
Loriaux DL (2012). An approach to the patient with hirsutism. JCEM 97, 2957–68.
Martin KA, Chang JR, Ehrmann DA, et al. (2008). Evaluation and treatment of hirsutism in premeno-
pausal women: an Endocrine Society clinical practice guideline. JCEM 93, 1105–20.
308 CHAPTER 4 Reproductive endocrinology
Pituitary
i Insulin i LH secretion
Adrenal
i Androstenedione i Androstenedione
Ovary i Testosterone i DHEAS
Obesity
Hyperinsulinaemia ??
Genetic
susceptibility
d SHBG i Free testosterone
i Free oestradiol
Fig. 4.2 Abnormalities of hormone secretion in PCOS.
310 CHAPTER 4 Reproductive endocrinology
Hyperinsulinaemia
• Approximately 70% of ♀ with PCOS are insulin-resistant, depending
on the definition. The defect in insulin sensitivity appears to be
selective, mainly affecting the metabolic effects of insulin (effects
on muscle and liver) but sparing the ovaries where insulin acts as a
co-gonadotrophin to amplify LH-mediated testosterone synthesis.
Hyperinsulinaemia is exacerbated by obesity but can also be present in
lean ♀ with PCOS.
• There is also evidence in a number of ♀ with PCOS of insufficient
B-cell response to a glucose challenge, which is known to be a
precursor to type 2 diabetes mellitus.
• Insulin also inhibits SHBG synthesis by the liver, with a consequent rise
in free androgen levels.
Features
• Onset of symptoms. Symptoms often begin around puberty, after weight
gain, or after stopping the oral contraceptive pill but can present at
any time.
• Oligo-/amenorrhoea (70%). Due to anovulation. ♀ are usually well
oestrogenized, so there is little risk of osteoporosis, unlike other
causes of amenorrhoea.
• Hirsutism (66%):
• 25% of ♀ also suffer from acne or male pattern alopecia. Virilization
is not a feature of PCOS.
• There is often a family history of hirsutism or irregular periods.
• Slower onset of hirsutism makes a distinction from adrenal
or ovarian tumours which are rapidly progressive and more
likely to be associated with virilization and higher testosterone
concentrations.
• <1% of hirsute ♀ have non-classic congenital adrenal hyperplasia
(b see Clinical presentation, p. 321), and this should be excluded,
particularly in ♀ with significantly raised serum testosterone levels.
• Obesity (50%). Symptoms worsen with obesity, as it is accompanied
by i testosterone concentrations as a result of hyperinsulinaemia.
Acanthosis nigricans may be found in 1–3% of insulin-resistant ♀
with PCOS.
• Infertility (30%). PCOS accounts for 75% of cases of anovulatory
infertility. The risk of spontaneous miscarriage is also thought to be
higher than the general population, mainly because of obesity.
POLYCYSTIC OVARY SYNDROME (PCOS) 311
Investigations of PCOS
The aims of investigations are mainly to exclude serious underlying disor-
ders and to screen for complications, as the diagnosis is primarily clinical
(see Box 4.3).
Confirmation of diagnosis
• Testosterone concentration:
• Performed primarily as a screen for the presence of other causes of
hyperandrogenism.
• Often normal in ♀ with PCOS, and serum androgen concentrations
do not reflect the degree of hirsutism because of variable androgen
receptor sensitivity.
• LH concentration:
• The higher the LH level, the more likely the risk of anovulation and
infertility.
• Cannot be used to diagnose PCOS, as it is normal in many affected
♀. Similarly, the traditional LH:FSH ratio is not a useful indicator.
• SHBG:
• Low in 50% of ♀ with PCOS, owing to the hyperinsulinaemic state,
with a consequent increase in circulating free androgens.
• Useful indirect marker of insulin resistance.
• Free androgen index: FAI = 100 x (total testosterone/SHBG).
• Anti-Müllerian hormone (AMH) made by preantral follicles raised in
PCOS but overlaps with normal range.
• Pelvic US of ovaries and endometrium:
• Ultrasound criteria for PCO defined >12 follicles between 2–9mm
in diameter or ovarian volume >10cm3.
• US is sensitive but not specific (occurs in CAH and
Cushing’s). Usually transvaginal but transabdominal possible in
experienced hands.
• False –ve results common in non-specialist scans.
• Measurement of endometrial thickness is of major importance
in the diagnosis of endometrial hyperplasia in the presence of
anovulation. Endometrial hyperplasia is diagnosed if the endometrial
thickness is >10mm.
• Transvaginal US will also identify 90% of ovarian virilizing tumours.
INVESTIGATIONS OF PCOS 313
Associated endocrinopathy
• Serum prolactin. In the presence of infertility or oligoamenorrhoea.
• Mild hyperprolactinaemia (up to 2000mU/L) is present in up to 30%
of ♀ with PCOS, and dopamine agonist treatment of this may be
necessary if pregnancy is desired.
• 17OH progesterone (17OHP) level:
• Used to exclude late-onset congenital adrenal hyperplasia.
• Indicated in those with testosterone concentrations in excess of
5nmol/L or with evidence of virilization.
• May also perform a Synacthen test, looking for an exaggerated
®
Management of PCOS
(See Table 4.2.)
Weight loss
Studies have uniformly shown that weight reduction in obese ♀ with
PCOS will improve insulin sensitivity and significantly reduce hyperan-
drogenaemia. Obese ♀ are less likely to respond to antiandrogens and
infertility treatment. With a loss of 5%, ♀ with PCOS show improvement
in hirsutism, restoration of menstrual regularity, and fertility.
Metformin
In obese and lean insulin-resistant ♀ with PCOS, metformin (1g–2.5g
daily) improves insulin sensitivity, with a corresponding reduction in
serum androgen and LH concentrations and an increase in SHBG levels.
Metformin may regulate menstruation by improving ovulatory function but
improved live birth rate not established. Metformin does not seem to
improve response rates to ovulation induction using clomifene or gon-
adotrophins. Some benefits of metformin may be related to other lifestyle
measures, such as diet and weight loss. Metformin is usually stopped at
the diagnosis of pregnancy, as benefit in pregnancy is not proved outside
of established GDM.
There have been few long-term studies looking at the effect of met-
formin on hirsutism, but it appears that its effects are modest at best, and
most ♀ with significant hirsutism will require an antiandrogen.
♀ should be warned of its gastrointestinal side effects. In order to
minimize these, they should be started on a low dose (500mg once daily)
which may be i gradually to a therapeutic dose over a number of weeks.
Hirsutism
Pharmacological treatment of hirsutism (see Table 4.2) is directed at slow-
ing the growth of new hair. It can be combined with mechanical methods
of hair removal, such as electrolysis and laser therapy. Therapy is most
effective when started early. There is slow improvement over the first
6–12 months of treatment. Patients should be warned that facial hair is
slow to respond, treatment is prolonged, and symptoms may recur after
discontinuation of drugs. Adequate contraception is mandatory during
pharmacological treatment of hirsutism because of possible teratogenicity.
MANAGEMENT OF PCOS 315
Infertility
Ovulation induction regimens are indicated. Obesity adversely affects fer-
tility outcome, with poorer pregnancy rates and higher rates of miscar-
riage, so weight reduction should be strongly encouraged.
Metformin
• Use remains controversial but probably does not improve pregnancy
rates in insulin-resistant, particularly overweight, ♀ with PCOS. No
reported teratogenic or neonatal complications.
• Dose. 500mg od after meals, to be i gradually to 1g bd. If ovulation
restored following 6 months of treatment, then continue for up to
1 year. If pregnancy does not occur, then consider other treatments.
• Side effects. Nausea, bloating, diarrhoea, vomiting.
Clomifene citrate
• Inhibits oestrogen negative feedback, i FSH secretion and thus
stimulating ovarian follicular growth.
• Dose. 25–150mg a day from day 2 of menstrual cycle for 5 days.
• Response rates. 80% ovulation rate, 67% pregnancy rate.
• Complications. 8% twins, 0.1% higher order multiple pregnancy. Risk of
ovarian neoplasia following prolonged clomiphene treatment remains
unclear, so limit treatment to a maximum of six cycles.
Gonadotrophin preparations (hMG or FSH)
• Used in those unresponsive to clomifene. Low-dose regimes show
better response rates and fewer complications, e.g. 75IU/day for
2 weeks, then increase by 37.5IU/day every 7 days, as required.
• 94% ovulation rate and 50% pregnancy rate.
• Complications. Hyperstimulation, multiple pregnancies.
• Close ultrasonic monitoring is essential.
Surgery
• Laparoscopic ovarian diathermy or laser drilling may restore ovulation
in up to 90% of ♀, with cumulative pregnancy rates of 80% within
8 months of treatment. Particularly effective in slim ♀ with PCOS and
high LH concentrations.
• Complications. Surgical adhesions although usually mild.
In vitro fertilization
• In ♀ who fail to respond to ovulation induction.
• 60–80% conception rate after six cycles.
Acne
Treatment for acne should be started as early as possible to prevent scar-
ring. All treatments take up to 12 weeks before significant improvement
is seen. All treatments, apart from benzoyl peroxide, are contraindicated
in pregnancy.
Mild-to-moderate acne
Topical benzoyl peroxide 5%. Bactericidal properties. May use in conjunc-
tion with oral antibiotic therapy to reduce the risk of developing resistance
to antibiotics. Side effects: skin irritation and dryness. Add oral antibiotics
if no improvement after 2 months of treatment.
318 CHAPTER 4 Reproductive endocrinology
Moderate-to-severe acne
• Topical retinoids, e.g. tretinoin, isotretinoin. Useful alone in mild acne
or in conjunction with antibiotics in moderately severe acne. Continue
as maintenance therapy to prevent further acne outbreaks. Side
effects: irritation, photosensitivity. Apply high factor sunscreen before
sun exposure. Avoid in acne involving large areas of skin.
• Oral antibiotics, e.g. oxytetracycline 500mg bd, doxycycline 100mg od, or
minocycline 100mg od. Response usually seen by 6 weeks and full efficacy
by 3 months. Continue antibiotics for 2 months after control is achieved.
Prescription usually given for a course of 3–6 months. Continue topical
retinoids and/or benzoyl peroxide to prevent further outbreaks.
• COCP and antiandrogens. As for hirsutism.
Severe acne
Isotretinoin is very effective in ♀ with severe acne or acne which has not
responded to other oral or topical treatments. Used early, it can minimize
scarring in inflammatory acne. However, it is highly toxic and can only be
prescribed by a consultant dermatologist. Also consider referring ♀ who
develop acne in their 30s or 40s and ♀ with psychological problems as a
result of acne.
Further reading
Diamanti-Kandarakis E, Dunaif A. (2012). Insulin resistance and the polycystic ovary syndrome
revisited: an update on mechanisms and implications. Endocr Rev 33, 981–1030.
Dunaif A (1997). Insulin resistance and polycystic ovary syndrome: mechanism and implications for
pathogenesis. Endoc Rev 18, 774–800.
Ehrmann DA (2005). Polycystic ovary syndrome. N Engl J Med 352, 1223–36.
Ehrmann DA, Rychlik D (2003). Pharmacological treatment of polycystic ovary syndrome. Semin
Reprod Med 21, 277–83.
Ledger WL, Clark T (2003). Long term consequences of polycystic ovary syndrome. Royal College of
Obsterics and Gynaecology Guideline number 33. Royal College of Obstetrics and Gynaecology,
London.
Lord JM, Flight IHK, Norman RJ (2003). Insulin sensitizing drugs for polycystic ovary syndrome.
Cochrane Database Syst review 2003.
Neithardt AB, Barnes RB (2003). The diagnosis and management of hirsutism. Semin Reprod Med
21,285–93.
Nestler JE (2008). Metformin for the treatment of the polycystic ovary syndrome. N Engl J Med
358, 47–54.
Palomba S (2009). Evidence-based and potential benefits of metformin in the polycystic ovary
syndrome: a comprehensive review. Endocr Rev 30, 1–50.
Paradisi R (2010). Retrospective observational study on the effects and tolerability of flutamide
in a large population of patients with various kinds of hirsutism over a 15-year period. Eur J
Endocrinol 163, 139–4.
Pierpoint T, McKeigue PM, Isaacs AJ, et al. (1998). Mortality of women with polycystic ovary
syndrome at long term follow up. J Clin Epidemiol 51, 581–6.
Randeva HS, et al. (2012). Cardiometabolic aspects of the polycystic ovary syndrome. Endocr Rev
33, 812–41.
The Rotterdam ESHRE/ASRM-sponsored PCOS consensus workshop group (2004). Revised
2003 Consensus on diagnostic criteria and long-term health risks related to polycystic ovary
syndrome. Hum Reprod 19, 41–7.
Tsilchorozidou T, Overton C, Conway GS (2004). The pathophysiology of polycystic ovary
syndrome. Clin Endocrinol 60, 1–17.
MANAGEMENT OF PCOS 319
320 CHAPTER 4 Reproductive endocrinology
Cholesterol
17α OH 17α OH
Pregnenolone 17OH Pregnenolone Dehydroepiandrosterone
3βHSD 17β HSD Aromatase
Progesterone 17OH Progesterone Androstenedione Testosterone Oestradiol
21 OH 21 OH
Deoxycorticosterone 11-Deoxycortisol
11βOH 11β OH
Corticosterone Cortisol
18-OH Corticosterone
Aldosterone
Clinical presentation
Classic CAH
• Most patients are diagnosed in infancy, and their clinical presentation is
discussed elsewhere (b see Congenital adrenal hyperplasia, p. 320).
• Problems persisting into adulthood. Sexual dysfunction and subfertility
in ♀, particularly in salt wasters. Reconstructive genital surgery is
required in the majority of ♀ who were virilized at birth to create an
adequate vaginal introitus. With improvement of medical care, normal
pregnancy rates (90%) can be achieved.
• In ♂, high levels of adrenal androgens suppress gonadotrophins
and thus testicular function. i ACTH results in the development of
testicular adrenal rest tissue (TARTs). These are always benign but
may be misdiagnosed as testicular tumours. TARTs can be destructive,
leading to testicular failure. Spermatogenesis is often low if CAH is
poorly controlled.
• There is a significant risk of adrenal crises over lifetime.
• There is an impaired quality of life.
Non-classic CAH
• Due to partial deficiency of 21A-hydroxylase. Glucocorticoid and
aldosterone production are normal, but there is overproduction of
17OHP and thus androgens.
• Present with hirsutism (60%), acne (33%), and oligomenorrhoea (54%),
often around the onset of puberty. Only 13% of ♀ present with
subfertility.
• Polycystic ovaries on US are common, and adrenal incidentalomas or
hyperplasia are seen in 40%.
• Asymptomatic in ♂. The effect of non-classic CAH on ♂ fertility is
unknown.
322 CHAPTER 4 Reproductive endocrinology
Monitoring of treatment
Annual follow-up is usually adequate in adults.
• Clinical assessment. Measure BP and weight. Look for evidence of
hyperandrogenism and glucocorticoid excess. Amenorrhoea in ♀
usually suggests inadequate therapy but may have to be accepted as
can often not be achieved without excessive doses of glucocorticoid.
• Suppressed levels of 17OHP, testosterone (T), and renin are an
indication for reduced dose of steroid. Normalizing will often result in
complications from supraphysiological doses of glucocorticoids.
• Modestly raised or high normal levels of 17OHP, T, and renin are
optimal.
• Consider bone density which may be reduced by supraphysiological
steroid doses.
• Testicular function requires special attention. Low LH is a sign of
raised adrenal androgens. Raised FSH may follow adrenal rests as a
sign of testicular failure. US testis every 3–5 years. Consider sperm
count/storage if adrenal rests are present.
• US ovaries not useful routinely, as PCO morphology is common.
• In men, testicular adrenal rest tumours may develop (up to 94%). Their
detection may prevent infertility later. Ultrasound screening of the
scrotum is suggested every 2 years.
Prognosis
Adults with treated CAH have a normal life expectancy. Improvement
in medical and surgical care has also improved QoL for most sufferers.
However, there are a few unresolved issues:
• Height. Despite optimal treatment in childhood, patients with CAH
are, on average, significantly shorter than their predicted genetic
height. Studies suggest that this may be due to overtreatment with
glucocorticoids during infancy.
• Fertility. Mainly a problem in ♂ with poorly controlled classic CAH
and adrenal rests.
• Adrenal incidentalomas. Benign adrenal adenomas have been reported
in up to 50% of patients with classic CAH. ♂ with CAH may develop
gonadal adrenocortical rests.
• Psychosexual issues. Gender dysphoria common in ♀. A significant
number of ♀ with classic CAH, despite adequacy of vaginal
reconstruction, are not sexually active.
MONITORING OF TREATMENT 327
Further reading
Arlt W, et al. (2010). Health status of adults with congenital adrenal hyperplasia: a cohort study of
203 patients. J Clin Endocrinol Metab 95, 5110–21.
Aycan Z, Bas VN, Cetinkaya S, et al. (2013) Prevalence and long-term follow-up outcomes of
testicular adrenal rest tumours in children and adolescent males with congenital adrenal hyper-
plasia. Clin Endocrinol 78, 667–72.
Cabrera MS, Vogiatzi MG, New MI (2001). Long term outcome in adult males with classic congeni-
tal adrenal hyperplasia. J Clin Endocrinol Metab 86, 3070–8.
Joint LWPES/ESPE CAH Working Group (2002). Consensus statement on 21-hydroxylase defi-
ciency from the Lawson Wilkins Paediatric Endocrine Society and the European Society for
Paediatric Endocrinology. J Clin Endocrinol Metab 87, 4048–53.
Merke DP (2008). Approach to the adult with congenital adrenal hyperplasia due to 21-hydroxylase
deficiency. JCEM 93, 653–60.
New MI (2006). Extensive clinical experience: nonclassical 21-hydroxylase deficiency. J Clin
Endocrinol Metab 91, 205–14.
New MI, Carlson A, Obeid J, et al. (2001). Prenatal diagnosis for congenital adrenal hyperplasia in
532 pregnancies. J Clin Endocrinol Metab 86, 5651–757.
Ogilvie CM, Crouch NS, Rumsby G, et al. (2006). Congenital adrenal hyperplasia in adults: a review
of medical, surgical and psychological issues. Clin Endocrinol 64, 2–11.
Premawaradhana LDKE, Hughes IA, Read GF, et al. (1997). Longer term outcome in females with
congenital adrenal hyperplasia: the Cardiff experience. Clin Endocrinol 46, 327–32.
Speiser PW, White PC (2003). Congenital adrenal hyperplasia. N Engl J Med 349, 776–88.
Speiser PW, et al. (2010). Congenital adrenal hyperplasia due to steroid 21-hydroxylase defi-
ciency: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 95, 4133–60.
328 CHAPTER 4 Reproductive endocrinology
Androgen-secreting tumours
Definition
Rare tumours of the ovary or adrenal gland which may be benign or malig-
nant, which cause virilization in ♀ through androgen production.
Epidemiology and pathology
Androgen-secreting ovarian tumours
• 75% develop before the age of 40 years.
• Account for 0.4% of all ovarian tumours; 20% are malignant.
• Tumours are 5–25cm in size. The larger they are, the more likely they
are to be malignant. They are rarely bilateral.
• Two major types:
• Sex cord stromal cell tumours: often contain testicular cell types.
• Adrenal-like tumours: often contain adrenocortical or Leydig cells.
• Other tumours, e.g. gonadoblastomas and teratomas, may also, on
occasion, present with virilization.
Androgen-secreting adrenal tumours
• 50% develop before the age of 50 years.
• Larger tumours, particularly >6cm, are more likely to be malignant.
• Usually with concomitant cortisol secretion as a variant of Cushing’s
syndrome.
Clinical features
• Onset of symptoms. Usually recent onset of rapidly progressive
symptoms.
• Hyperandrogenism:
• Hirsutism of varying degree, often severe; male pattern balding and
acne are also common.
• Usually oligo-/amenorrhoea.
• Infertility may be a presenting feature.
• Virilization. (b see Box 4.1, p. 305.) Indicates severe hyperandrogenism,
is associated with clitoromegaly, and is present in 98% of ♀ with
androgen-producing tumours. Not usually a feature of PCOS.
• Other:
• Abdominal pain.
• Palpable abdominal mass.
• Ascites.
• Symptoms and signs of Cushing’s syndrome are present in many of
♀ with adrenal tumours.
Investigations
See Fig. 4.4.
ANDROGEN-SECRETING TUMOURS 329
Serum testosterone
>5 i nmol/L
Ovarian US
MRI adrenal glands and ovaries
Management
Surgery
• Adrenalectomy or ovarian cystectomy/oophorectomy.
• Curative in benign lesions.
Adjunctive therapy
Malignant ovarian and adrenal androgen-secreting tumours are usually
resistant to chemotherapy and radiotherapy.
Prognosis
Benign tumours
• Prognosis excellent.
• Hirsutism improves post-operatively, but clitoromegaly, male pattern
balding, and deep voice may persist.
Malignant tumours
• Adrenal tumours. 20% 5-year survival. Most have metastatic disease at
the time of surgery.
• Ovarian tumours. 30% disease-free survival and 40% overall survival at
5 years.
Further reading
Hamilton-Fairley D, Franks S (1997). Androgen-secreting tumours. In: Sheaves R, Jenkins PJ, Wass
JAH (eds.) Clinical endocrine oncology, pp. 323–9. Blackwell Science, Oxford.
Rothman MS, Wierman ME (2011). How should postmenopausal androgen excess be evaluated?
Clin Endocrinol (Oxf) 75, 160–4.
330 CHAPTER 4 Reproductive endocrinology
Menstrual function
disorder—investigations
(See Fig. 4.5.)
• Is it p or s ovarian dysfunction?
• FSH, LH, TFT, oestradiol, prolactin.
• Ultrasound:
• Ovarian and uterine morphology—exclude anatomical
abnormalities, PCOS, and Turner’s syndrome.
• Note that PCO morphology is present in 20% of all women, so
their presence may be false +ve.
• Endometrial thickness—to assess oestrogen status.
• Other tests, depending on clinical suspicion:
• Induce withdrawal bleed with progesterone (e.g. 10mg
medroxyprogesterone acetate bd for 7 days). If a bleed occurs,
then there is adequate oestrogen priming and endometrial
development. This test has poor specificity and sensitivity and has
been abandoned in favour of ultrasound in many centres.
• Serum testosterone in the presence of hyperandrogenism.
• Karyotype in ovarian failure or disorder of sexual development
suspects (absent uterus).
• MRI of the pituitary fossa if FSH low or in the presence of
hyperprolactinaemia.
• Bone density if long-term oestrogen deficiency.
Exclude
pregnancy
FSH, LH,
prolactin
1º Hypogonadotrophic Hyperprolactinaemia:
ovarian failure: hypogonadism: rule out PCOS
rule out hypothyroidism and
check karyotype pituitary adenoma pituitary adenoma
Management of amenorrhoea
• Treat underlying disorder, for example:
• Dopamine agonists for prolactinomas.
• Pituitary surgery for pituitary tumours.
• Eating disorder clinic in anorexia nervosa.
• Treat oestrogen deficiency—oestrogen/progestagen preparations.
• Treat infertility—b see p. 334.
• In PCOS with endometrial hyperplasia—progesterone withdrawal
bleed, and consider hysteroscopy in resistant cases.
• See Box 4.6 for progesterone sensitivity.
Further reading
Baird DT (1997). Amenorrhoea. Lancet 350, 275–9.
Beswick SJ, Lewis HM, Stewart PM (2002). A recurrent rash treated by oophorectomy. QJM
95, 636–7.
Hickey M, Balen A (2003). Menstrual disorders in adolescence: investigation and management. Hum
Reprod Update 9, 493–504.
336 CHAPTER 4 Reproductive endocrinology
Pathogenesis
POI is the result of accelerated depletion of ovarian germ cells. Previously
used term of ‘resistant ovary syndrome’ describes a mild form of POI that
gradually progresses to amenorrhoea. Ovarian biopsy is no longer used,
as this had no diagnostic value. Conversely, ovarian dysgenesis refers to
early onset with primary amenorrhoea and describes the severe end of
the condition.
POI is usually permanent and progressive, although a remitting course is
also experienced and cannot be fully predicted, so all women must know
that pregnancy is possible, even though fertility treatments are not effec-
tive (often a difficult paradox to describe). Spontaneous pregnancy has
been reported in 5%.
PREMATURE OVARIAN INSUFFICIENCY (POI) 337
Clinical presentation
• Amenorrhoea:
• May be p or s.
• Symptoms of oestrogen deficiency:
• Not present in those with p amenorrhoea.
• 75% of ♀ who develop s amenorrhoea report hot flushes, night
sweats, mood changes, fatigue, or dyspareunia; symptoms may
precede the onset of menstrual disturbances.
• Autoimmune disease:
• Screen for symptoms and signs of associated autoimmune
disorders.
• Other:
• Past history of radiotherapy, chemotherapy, or pelvic surgery.
• +ve family history in 20% of patients; careful history is required to
detect inheritance through father.
• With idiopathic POI, ovarian function can resume in most within a
year of diagnosis. Spontaneous pregnancies have been recorded, so
ultrasound assessment of follicles and measurement of inhibin B and
oestradiol may be helpful.
• See Box 4.8 for autoimmune diseases.
Investigation of POI
• Serum gonadotrophins:
• Diagnosis is confirmed by serum FSH >40mIU/L on at least two
occasions at least 1 month apart.
• Disease may have a fluctuating course, with high FSH levels
returning to normal and later regain of ovulatory function.
• LH also elevated but FSH usually disproportionately higher
than LH.
• Serum oestradiol levels are usually low.
• Anti-Müllerian hormone—only useful in early stages or at-risk groups,
not in established cases.
• Karyotype:
• All ♀ presenting with hypergonadotrophic amenorrhoea below age
40 should be karyotyped.
• ♀ with Y chromosomal material should be referred for bilateral
gonadectomy to prevent the development of gonadoblastoma.
• Pelvic US—to identify normal ovarian and uterine morphology.
• Bone mineral density—risk of osteoporosis.
• Screen for autoimmune disease:
• Thyroid autoantibodies: if +ve, extend autoimmune screen to
include adrenal, parietal cell, intrinsic factor.
• Ovarian antibodies are rarely +ve—poor sensitivity.
• TSH, vitamin B12.
• Synacthen test only if adrenal insufficiency is suspected clinically.
®
Management of POI
Sex hormone replacement therapy
• Exogenous oestrogens (HRT) are required to alleviate symptoms
and prevent the long-term complications of oestrogen deficiency—
osteoporosis and possibly cardiovascular disease. Initial doses depend
on the duration of amenorrhoea—if oestrogen-deficient for at least
12 months, then start on lowest doses of estradiol available to prevent
side effects, but titrate up to full dose within 6 months. If recently
amenorrhoeic, then full dose may be commenced immediately (see
Table 4.7).
• Doses used in HRT are not contraceptive and do not suppress
spontaneous ovarian follicular activity. Women who do not wish
to conceive may use COCP instead of HRT—beware E2-deficient
symptoms in pill-free week.
• HRT should be continued at least until the age of 50 years, the mean
age of the natural menopause.
• In non-hysterectomized ♀, a progestagen should be added for
12–14 days a month to prevent endometrial hyperplasia.
• Low-dose androgen replacement therapy may improve persistent
fatigue and poor libido, despite adequate oestrogen replacement.
Fertility
• A minority of ♀ with POI and a normal karyotype will recover
spontaneously; 5% spontaneous fertility rate.
• Ovum donation is the only realistic chance of fertility. Pregnancy rate
>50% per patient. Results are less good in ♀ if uterine damage after
pelvic radiotherapy.
• Ovulation induction therapy has been tried, but the results have
been poor.
• Glucocorticoid therapy has been used in autoimmune POI, but efficacy
is poor.
• Recent improvements in methods of oocyte cryopreservation using
rapid freezing in liquid nitrogen (vitrification) have allowed ♀ with high
chance of POI (e.g. before chemo-/radiotherapy, Turner’s syndrome
mosaics with retained ovarian function) to store oocytes collected
after superovulation.
Psychology
• Reduced self-esteem and sexual responsiveness.
• Reactive depression common at diagnosis and may return at critical
times, such as pregnancy in a close friend.
For a full review of hormone replacement therapy, b see Menopause,
p. 342.
MANAGEMENT OF POI 341
Prognosis
• Mortality of ♀ with POI may be i 2-fold.
• Oestrogen deficiency leads to:
• i risk of cardiovascular and cerebrovascular disease.
• i risk of osteoporosis. Up to two-thirds of ♀ with POI and a
normal karyotype have d BMD, with a Z score of –1 or less,
despite at least intermittent hormone replacement therapy.
This may be due to a combination of factors, including an initial
delay in initiating ERT, poor compliance with ERT, and oestrogen
‘underdosing’.
Further reading
Barlow DH (1996). Premature ovarian failure. Baillière Clin Ob Gy 10, 361–84.
Davies MC, Cartwright B (2012). What is the best management strategy for a 20-year-old woman
with premature ovarian failure? Clin Endocrinol (Oxf) 77, 182–6.
Kalantaridou SN, Davis SR, Nelson LM (1998). Premature ovarian failure. Endocrinol Metab Clin
27, 989–1006.
Nelson LM (2009). Clinical practice. Primary ovarian insufficiency. N Engl J Med 360, 606–14.
Welte CK (2008). Primary ovarian insufficiency: a more accurate term for premature ovarian
failure. Clin End 68, 499–509.
342 CHAPTER 4 Reproductive endocrinology
Menopause
Definition
• The menopause is the permanent cessation of menstruation as a result
of ovarian failure and is a retrospective diagnosis made after 12 months
of amenorrhoea. The average age of ♀ at the time of the menopause
is 750 years, although smokers reach the menopause 72 years earlier.
• The perimenopause encompasses the menopause transition and the
first year following the last menstrual period.
Physiology
• Ovaries have a finite number of germ cells, with maximal numbers at
20 weeks of intrauterine life. Thereafter, there is a gradual reduction
in the number of follicles until the perimenopause when there is an
exponential loss of oocytes until the store is depleted at the time of
the menopause.
• Inhibin B and anti-Müllerian hormone (AMH) are ovarian glycoproteins
produced by follicles, as they develop from preantral to antral stages.
Both may participate in ovarian paracrine regulation and, with other
molecules, regulate the rate of attrition of follicles.
• Falling inhibin B levels result in fluctuating rise in FSH. Contrary to
previous belief, average serum oestradiol levels may be high at the
onset of the menopause transition as a result of FSH rise, falling only
towards the end as the follicles are depleted.
• AMH is a useful early marker of ovarian ageing, being stable
throughout the cycle and unaffected by COCP and without the
marked fluctuations of FSH. AMH measurements, however, add
nothing in routine practice or established menopause.
• See Table 4.8 for hormonal changes.
Long-term consequences
• Osteoporosis. During the perimenopausal period, there is an
accelerated loss of bone mineral density (BMD), rendering
post-menopausal ♀ more susceptible to osteoporotic fractures.
• Ischaemic heart disease (IHD). Post-menopausal ♀ are 2–3x more likely
to develop IHD than premenopausal ♀, even after age adjustments.
The menopause is associated with an increase in risk factors for
atherosclerosis, including less favourable lipid profile, d insulin
sensitivity, and an i thrombotic tendency.
• Dementia.♀ are 2–3x more likely to develop Alzheimer’s disease than
♂. It is suggested that oestrogen deficiency may play a role in the
development of dementia.
Table 4.8 Hormonal changes during the menopausal transition
Premenopause Early perimenopause Advanced perimenopause Meno
(from age 36 years)
Menstrual Regular, ovulatory Irregular, often short cycles, Oligomenorrhoea Amen
cycle increasingly anovulatory
FSH Rising but within normal Intermittently raised, especially Persistently i ii
range in follicular phase
AMH Declining Low Low Very l
E2 Normal High normal Normal/low Low
344 CHAPTER 4 Reproductive endocrinology
Evaluation of menopause
(e.g. if HRT is being considered)
History
• Perimenopausal symptoms and their severity.
• Assess risk factors for cardiovascular disease and osteoporosis.
• Assess risk factors for breast cancer and thromboembolic disease.
• History of active liver disease.
Examination
• BP.
• Breasts.
• Consider pelvic examination for dyspareunia.
Investigations
• FSH levels fluctuate markedly in the perimenopausal period and
correlate poorly with symptoms. Remember, a raised FSH in the
perimenopausal period may not necessarily indicate infertility, so
contraception, if desired, should continue until the menopause.
• Mammography indicated prior to starting oestrogen replacement
therapy only in high-risk ♀; otherwise, mammography should be
offered as per national screening programme.
• Endometrial biopsy does not need to be performed routinely but is
essential in ♀ with abnormal uterine bleeding.
346 CHAPTER 4 Reproductive endocrinology
Hormone replacement
therapy (HRT)
The aim of treatment of perimenopausal ♀ is to alleviate menopausal
symptoms and optimize quality of life. The majority of women with mild
symptoms require no HRT. See Table 4.9 for alternatives to HRT.
Benefits of HRT
Hot flushes
Respond well to oestrogen therapy in a dose-dependent manner. Start
with a low dose, and increase gradually, as required, to control symptoms.
High doses may be required initially, particularly in younger ♀ or in those
whose symptoms develop abruptly (post-oophorectomy). In 75% of ♀,
vasomotor symptoms settle within 5 years, so consider stopping HRT
after 5 years of treatment. The dose of HRT should be gradually reduced
over weeks, as sudden withdrawal of oestrogen may precipitate the return
of vasomotor symptoms.
In ♀ with a contraindication to HRT or who are intolerant of it,
non-hormonal therapies are summarized in Table 4.9.
Urinary symptoms
A trial of HRT, local or systemic, may improve stress and urge inconti-
nence as well as the frequency of cystitis.
Vaginal atrophy
Systemic or local oestrogen therapy improves vaginal dryness and dys-
pareunia. A maximum of 6 months’ use of vaginal cream is recommended,
unless combined with a progestagen, as systemic absorption may increase
the risk of endometrial hyperplasia. If oestrogens are not successful, then
vaginal lubricants, e.g. Replens®, may help.
Osteoporosis
HRT has been shown to increase bone mineral density in the lumbar spine
by 3–5% and at the femoral neck by about 2% by inhibiting bone resorption.
The Women’s Health Initiative (WHI) trial confirmed that HRT reduces
the risk of both hip and vertebral fractures by 30%. HRT is not primarily
used for bone protection, as other agents may have fewer side effects
Colorectal cancer
WHI showed a 20% reduction in the incidence of colon cancer in HRT
users. However, HRT should currently not be prescribed solely to prevent
colorectal cancer.
Risks of HRT
(Also see Box 4.9.)
Breast cancer
No personal or family history of breast cancer
There is an i risk of breast cancer in HRT users which is related to the
duration of use. The risk increases by 35%, following 5 years of use (over
the age of 50), and falls to never-used risk 5 years after discontinuing HRT.
HORMONE REPLACEMENT THERAPY (HRT) 347
For ♀ aged 50 not using HRT, about 45 in every 1,000 will have cancer
diagnosed over the following 20 years. This number increases to 47/1,000
♀ using HRT for 5 years, 51/1,000 using HRT for 10 years, and 57/1,000
after 15 years of use. The risk is highest in ♀ on combined HRT compared
with oestradiol alone. Mortality has not been shown to be i in breast
cancer developing in ♀ on HRT.
Family history of breast cancer
The risk of breast cancer may be i 4-fold as a result of the family history,
but there is little evidence that the risk is i further by the use of HRT.
HRT may be used in these ♀ if severe vasomotor symptoms are present
after counselling regarding the above risks.
Past history of breast cancer
Avoid HRT in ♀ with a past history of breast cancer.
Venous thromboembolism (VTE)
Transdermal E2 is generally thought not to have an increased risk of VTE,
which is why this group is favoured in most situations.
Oral HRT increases the risk approximately 3-fold, resulting in an extra
two cases/10,000 woman-years. The risk is highest in the first year of use
of HRT and has been shown to be halved by aspirin or statin therapy. This
risk is markedly i in ♀ who already have risk factors for DVT, including
previous DVT, cardiovascular disease, and within 90 days of hospitalization.
Family history of DVT
If HRT is being considered because of severe vasomotor symptoms, then
do a thrombophilia screen. If +ve, then avoid HRT. ♀ with a +ve fam-
ily history of thromboembolism are still at a slightly i risk themselves,
even if the results of the thrombophilia screen are –ve, so consider
transdermal route.
Past history of DVT/PE
Risk of recurrence is 5% per year, so avoid HRT unless on long-term
warfarin therapy.
Cerebrovascular disease
HRT has been shown to increase the risk of ischaemic stroke in older ♀,
particularly in the presence of atrial fibrillation. HRT should, therefore, not
be used in ♀ with a history of cerebrovascular disease or atrial fibrillation,
unless they are anticoagulated.
Endometrial cancer
No i risk in ♀ taking continuous combined HRT preparations. ♀ using
sequential combined preparations do not appear to have an i risk of
endometrial cancer initially, but the risk of endometrial hyperplasia does
increase with long-term use (>5 years) despite regular withdrawal bleeds,
so these ♀ need regular follow-up. ♀ with cured stage I tumours may
safely take HRT.
Ovarian cancer
HRT may be associated with an i risk of ovarian cancer, but the evidence
is insufficient at present.
HORMONE REPLACEMENT THERAPY (HRT) 349
Gallstones
The risk of gallstones is i 2-fold in HRT users.
Migraine
Migraines may increase in severity and frequency in HRT users. A trial
of HRT is still worthwhile if indications are present, providing there are
no focal neurological signs associated with the migraine. Modification in
the dose of oestrogen or its preparation may improve symptoms. Avoid
conjugated oestrogens, as these are most commonly associated with an
increase in the frequency of migraines.
Endometriosis and uterine fibroids
The risk of recurrence of endometriosis or of growth of uterine fibroids
is low on HRT.
Liver disease
Use parenteral or transcutaneous oestrogens to avoid hepatic metabo-
lism, and monitor liver function in ♀ with impaired liver function tests. Do
not use HRT in the presence of active liver disease or liver failure.
Areas of uncertainty with HRT
Cardiovascular disease (CVD)
Data from >30 observational studies suggest that HRT may reduce the risk
of developing CVD by up to 50%. However, randomized placebo-controlled
trials, e.g. the Heart and Oestrogen-Progestin Replacement Study (HERS)
and the WHI trial, have failed to show that HRT protects against IHD.
Currently, HRT should not be prescribed to prevent cardiovascular dis-
ease. However, it may be used cautiously in individual patients with CVD
if QoL is significantly reduced from vasomotor symptoms.
Alzheimer’s disease
Recent evidence suggests that the risk of developing Alzheimer’s disease
may be reduced by up to 50% in ♀ receiving HRT, particularly if started
early in the menopause. However, in ♀ with established Alzheimer’s
disease, there is no evidence to suggest reversal of cognitive dysfunc-
tion following initiation of HRT. There is currently insufficient evidence
to recommend the use of HRT to prevent Alzheimer’s disease. In
post-menopausal ♀ without dementia, HRT may improve certain aspects
of cognitive function.
Mood disturbances
There has been a strongly held belief that HRT improves well-being and
QoL in perimenopausal and post-menopausal ♀. However, in recent tri-
als where QoL has been assessed, notably HERS and WHI, the improve-
ment in QoL and improved sleep was only seen in ♀ with vasomotor
symptoms.
350 CHAPTER 4 Reproductive endocrinology
Table 4.10 Risks and benefits per 10,000 ♀ treated with HRT per
year (WHI trial)
Benefits Number of patients
Hip fractures prevented 5
Colon cancer prevented 6
Adverse events:
Coronary heart disease 7
Cerebrovascular events 8
Pulmonary embolism 5
Breast cancer (>5 years’ use) 8
352 CHAPTER 4 Reproductive endocrinology
HRT regimens
Oestrogen preparations
See Table 4.11. In younger, symptomatic, often perimenopausal ♀, higher
doses of oestrogen are often required initially, which can be reduced grad-
ually to the lowest dose effective at controlling symptoms.
Older ♀ who have been amenorrhoeic for over a year should be
started on the lowest possible dose of oestrogen.
Route of administration
• Oral route is the most popular. Disadvantages:
• First pass hepatic metabolism results in increased thrombotic risk.
• May be associated with nausea and may exacerbate liver disease.
• Must be taken daily, so there is no breakthrough of symptoms.
• Transdermal patches avoid first pass effect and are thus ideal in ♀ with
liver disease or hypertriglyceridaemia. Additionally, patches provide
constant systemic hormone levels. However, 10% of ♀ develop skin
reactions. Try to avoid moisture and to rotate patch sites to prevent this.
• Gels have the advantages of patches, but skin irritation is less common.
• SC implants have the advantage of good compliance. However, if
side effects develop, implants are difficult to remove. Additionally,
may release oestradiol for up to 3 years after insertion, and cyclical
progestagens must be given until oestrogen levels are not detectable.
Progestagen preparations
See Table 4.12. Must be added in non-hysterectomized ♀ to avoid endo-
metrial hyperplasia and subsequent carcinoma.
Sequential cyclical regimen
Give progestagen for a minimum of 10 days a month. Usually given for
the first 12 days of each calendar month. Quarterly regimen available—
progestagen given for 14 days 4x a year. However, the risk of endometrial
hyperplasia on such a regimen is unknown.
99% of ♀ have a monthly withdrawal bleed. 10% may be amenorrhoeic,
with no harmful consequences. Bleeding should start after the ninth day
of progestagen therapy.
Continuous combined regimen
Lower doses of progestagen are given on a daily basis. Uterine bleeding is
usually light in amount, but timing is unpredictable. Bleeding should stop in
90% of ♀ within 12 months, the majority in 6 months.
Ideal for older ♀ who do not want monthly withdrawal bleeds.
Contraindicated in perimenopausal ♀, as irregular uterine bleeding is
more likely and difficult to assess.
Tibolone (2.5mg a day)
A synthetic steroid with mixed oestrogenic, progestagenic, and weak andro-
genic activities. An alternative form of HRT, with little stimulation of the
endometrium. It alleviates vasomotor symptoms, may improve mood and
libido, and is protective against osteoporosis. Risks of VTE and breast cancer
similar to oestrogen. 10% of ♀ may experience vaginal bleeding on tibolone.
HRT REGIMENS 353
Further reading
Arlt W (2006). Androgen therapy in women. Eur J Endocrinol 154, 1–11.
Davis SR, et al. (2008). Testosterone for low libido in postmenopausal women not taking estrogen.
N Engl J Med 359, 2005–17.
Humphries KH, Gill S (2003). Risks and benefits of hormone replacement therapy: the evidence
speaks. Can Med Assoc J 168, 1001–10.
Million Women Study Collaborators (2003). Breast cancer and hormone-replacement therapy in
the Million women Study. Lancet 362, 419–23.
Rymer J, Wilson R, Ballard K (2003). Making decisions about hormone replacement therapy. BMJ
326, 322–6.
Writing Group for Women’s Health Initiative Investigators (2002). Risks and benefits of estrogen
plus progestin in healthy postmenopausal women: principal results from the Women’s Health
Initiative randomized controlled trial. JAMA 288, 321–33.
HORMONAL CONTROL OF CONTRACEPTION 355
Venous thromboembolism
• The risk of venous thromboembolism in non-pregnant ♀ (5 per
100,000 ♀/year) is i 3-fold in ♀ on the COCP.
• The risk is highest in the first year of use, increases with age, and is i
in obese ♀.
• The risk of venous thromboembolism appears to be higher in ♀ taking
COCPs containing desogestrel, gestodene, cyproterone acetate, or
drospirenone, but absolute risk may be small in those with low risk
and outweighed by benefits.
• ♀ with a family history of thromboembolism should undergo a
thrombophilia screen before starting the COCP.
Arterial thrombosis
• There is a 10-fold excess risk of IHD in ♀ smokers over the age of
35 years who are on the COCP. The risk of IHD does not seem to be
significantly i in non-smokers who take low-dose COCP.
• The relative risk of ischaemic stroke is only slightly i in ♀ taking
low-dose COCP. The risk is i in ♀ over the age of 35 years, smokers,
or in ♀ with hypertension. The risk of haemorrhagic stroke does not
seem to be i by taking the COCP.
• Risk of either arterial or venous thrombosis returns to normal within
3 months of discontinuing the COCP.
Hypertension
May be caused by the COCP and, if already present, may be more resist-
ant to treatment.
Hepatic disease
Raised hepatic enzymes may be seen in ♀ on the COCP. The incidence of
benign hepatic tumours is also i.
Gallstones
The risk of developing gallstones is slightly i by taking the COCP
(RR = 1.2).
Breast cancer
• There may be a slightly i risk of breast cancer in ♀ using the COCP
(RR = 1.3), particularly in those who began taking the COCP in
their teens.
• The risk does not seem to be related to the duration of exposure to
the COCP nor to the EE2 dose.
• The relative risk of developing breast cancer returns to normal
10 years after discontinuing the COCP.
Cervical cancer
• The use of COCP appears to be associated with an excess risk of
cervical cancer in ♀ who are HPV +ve. The risk increases with the
duration of COCP use.
• It is not known whether the risk falls again following the
discontinuation of COCP.
COMBINED ORAL CONTRACEPTIVE PILLS (COCP) 359
Thrombophilia screen
• Antithrombin III.
• Protein C.
• Protein S
• Factor V Leiden.
Practical issues
Age and the COCP
• ♀ with no risk factors for arterial or venous thrombosis may continue
to use the combined COCP until the age of 50 years.
• Those with risk factors for thromboembolism and IHD should avoid
COCP after the age of 35 years, particularly if they are smokers.
• Contraception after the menopause: assume fertile for the first year
after last menstrual period if >50 years.
Breakthrough bleeding
Causes include:
• Genital tract disease.
• Insufficient oestrogen dose.
• Inappropriate progestagen.
• Missed pill.
• Taking two packets continuously.
• Gastroenteritis.
• Drug interactions, e.g. antibiotics, hepatic enzyme inducers.
Antibiotics and the COCP
Broad-spectrum antibiotics interfere with intestinal flora, thereby reducing
bioavailability of the COCP. Additional methods of contraception should
be used.
COCP and surgery
• Stop COCP at least 4 weeks before major surgery and any surgery to
the legs. Do not restart until fully mobile for at least 2 weeks.
• If emergency surgery, then stop COCP and start antithrombotic
prophylaxis.
360 CHAPTER 4 Reproductive endocrinology
Emergency contraception
Refers to contraception that a woman can use after unprotected sexual
intercourse to prevent pregnancy.
• A single 1.5mg dose of the progestagen levonorgestrel is highly
efficacious, with a pregnancy rate of 2–4% if taken within 72h of sexual
intercourse. It is most effective the earlier it is taken. The dose is
repeated if vomiting occurs within 3h of taking the pill.
• The mechanism of action is not clear but is thought to be due to
inhibition of ovulation as well as a d likelihood of endometrial
implantation.
• Side effects. Nausea in up to 60% of ♀, vomiting in 10–20%. Consider
antiemetic 1h before taking contraception. Other side effects—breast
tenderness, fatigue, dizziness.
• Contraindication—pregnancy.
• ♀ should be advised to use barrier contraception until their next
period.
• 98% of ♀ menstruate within 3–4 weeks of taking levonorgestrel. They
should be encouraged to seek medical advice if they do not bleed in
that time.
• If pregnancy does occur, levonorgestrel is not known to be
teratogenic.
Further reading
Lidegaard O (2012). Thrombotic stroke and myocardial infarction with hormonal contraception.
N Engl J Med. 366, 2257–66.
Petitti DB (2003). Combination oestrogen-progestin oral contraceptives. N Engl J Med 349, 1443–50.
MALE HYPOGONADISM 361
Male hypogonadism
Definition
Failure of testes to produce adequate amounts of testosterone, sperma-
tozoa, or both.
Epidemiology
• Klinefelter’s syndrome (XXY) (b see p. 370) is the most common
congenital cause and is thought to occur with an incidence of 2:1,000
live births.
• Acquired hypogonadism is even more common, affecting 1:200 ♂.
Evaluation of male hypogonadism
Presentation
• Failure to progress through puberty.
• Sexual dysfunction.
• Infertility.
• Non-specific symptoms, e.g. lethargy, reduced libido, mood changes,
weight gain.
The clinical presentation depends on:
• The age of onset (congenital vs acquired).
• The severity (complete vs partial).
• The duration (functional vs permanent).
362 CHAPTER 4 Reproductive endocrinology
Secondary hypogonadism
Definition
Hypogonadism as a result of hypothalamic or pituitary dysfunction.
Diagnosis
• Low 9 a.m. serum testosterone.
• Low normal or low LH and FSH, normal inhibin B and anti-Müllerian
hormone.
Causes
(See Box 4.15.)
Kallmann’s syndrome
A genetic disorder characterized by failure of episodic GnRH secretion 9
anosmia. Results from disordered migration of GnRH-producing neurons
into the hypothalamus.
Epidemiology
• Incidence of 1 in 10,000 ♂.
• ♂:♀ ratio = 4:1.
Diagnosis
• Anosmia in 75%.
• i risk of cleft lip and palate, sensorineural deafness, cerebellar ataxia,
and renal agenesis.
• Low testosterone, LH, and FSH levels.
• Rest of pituitary function normal.
• Normal MRI pituitary gland and hypothalamus; absent olfactory bulbs
may be seen on MRI.
• Normalization of pituitary and gonadal function in response to
physiological GnRH replacement.
Genetics
• Most commonly, a result of an isolated gene mutation.
• May be inherited in an X-linked (KAL1), autosomal dominant FGFR1
mutation/KAL2, or recessive trait.
• KAL1 gene mutation, responsible for some cases of X-linked
Kallmann’s syndrome, is located on Xp22.3. It has a more severe
reproductive phenotype.
• Mutations of FGFR1 (fibroblast growth factor receptor 1) gene, located
on chromosome 8p11, is associated with the autosomal dominant
form of Kallmann’s syndrome. Affected ♂ have an i likelihood of
undescended testes at birth.
• 12–15% incidence of delayed puberty in families of subjects with
Kallman’s syndrome, compared with 1% in general population.
• ♂ with autosomal dominant form (FGFR1 mutation)—50% transmitted
to offspring who have increased likelihood of undescended testes at birth.
Management
• Androgen replacement therapy.
• When fertility is desired, testosterone is stopped and exogenous
gonadotrophins are administered (b see p. 408).
SECONDARY HYPOGONADISM 363
Structural
• Usually associated with other pituitary hormonal deficiencies.
• In children, craniopharyngiomas are the most common cause. Cranial
irradiation for leukaemia or brain tumours may also result in s
hypogonadism.
• The commonest lesions in adulthood are prolactinomas.
Systemic illness
Severe illness of any kind may cause hypogonadotrophic hypogonadism
(see Box 4.14).
Prader–Willi syndrome
A congenital syndrome affecting 1:25,000 births, caused by loss of an
imprinted gene on paternally derived chromosome 15q11–13.
It should be suspected in infancy in the presence of characteristic facial
features (almond eyes, downturned mouth, strabismus, thin upper lip),
severe hypotonia, poor feeding, and developmental delay. The child then
develops hyperphagia due to hypothalamic dysfunction, resulting in severe
obesity. Other characteristic features include short stature, hypogonado-
trophic hypogonadism, and learning disability.
• Type 2 diabetes occurs in 15–40% of adults.
Laurence–Moon–Biedl syndrome
Congenital syndrome characterized by severe obesity, gonadotrophin
deficiency, retinal dystrophy, polydactyly, and learning disability.
CHARGE syndrome
The association of coloboma, heart anomaly, choanal atresia, retardation,
genital and ear anomalies. A proportion also has HH.
368 CHAPTER 4 Reproductive endocrinology
Primary hypogonadism
Due to testicular failure with normal hypothalamus and pituitary function.
See Box 4.17 for clinical characteristics.
Diagnosis
• Low 9 a.m. serum testosterone.
• Elevated LH and FSH, low inhibin B, and anti-Müllerian hormone.
Causes
Genetic
Klinefelter’s syndrome
The most common congenital form of p hypogonadism. It is thought that
a significant number of men with Klinefelter’s syndrome are not diagnosed.
Clinical manifestations will depend on the age of diagnosis. Patients with
mosaicism tend to have less severe clinical features.
• Adolescence:
• Small firm testes (mean 5mL).
• Gynaecomastia.
• Tall stature (i leg length).
• Other features of hypogonadism.
• Cognitive dysfunction.
• Adulthood:
• Reduced libido and erectile dysfunction.
• Gynaecomastia (50%).
• Reduced facial hair.
• Obesity.
• Infertility.
• Risks:
• Type 2 diabetes mellitus.
• Osteoporosis.
• Thromboembolism.
• Malignancies, e.g. extragonadal germ cell tumours.
• Diagnosis:
• Karyotyping: 47,XXY in 80%; higher grade chromosomal
aneuploidies (e.g. 48,XXXY) or 46,XY/47,XXY mosaicism in the
remainder.
• Low testosterone, elevated FSH and LH.
• Elevated SHBG and oestradiol.
• Azoospermia.
• Management:
• Lifelong androgen replacement.
• May need surgical reduction of gynaecomastia.
• Fertility: intracytoplasmic sperm injection (ICSI, b see Management
of male infertility, p. 409), using testicular spermatozoa from men
with Klinefelter’s syndrome, has resulted in successful pregnancies.
However, couples should be counselled about the i risk of
chromosomal abnormalities in offspring.
PRIMARY HYPOGONADISM 371
Clinical assessment of
hypogonadism
History
• Developmental history. Congenital urinary tract abnormalities, e.g.
hypospadias, late testicular descent, or cryptorchidism.
• Delayed or incomplete puberty.
• Infections, e.g. mumps, orchitis.
• Abdominal/genital trauma.
• Testicular torsion.
• Anosmia.
• Drug history, e.g. sulfasalazine, antihypertensives, chemotherapy,
cimetidine, radiotherapy; alcohol and recreational drugs also
important.
• General medical history. Chronic illness, particularly respiratory,
neurological, and cardiac.
• Gynaecomastia. Common (b see Gynaecomastia, pp. 382–3) during
adolescence. Recent-onset gynaecomastia in adulthood—must rule out
oestrogen-producing tumour.
• Family history. Young’s syndrome (reduced fertility, bronchiectasis, and
rhinosinusitis), cystic fibrosis, Kallmann’s syndrome.
• Sexual history. Erectile function, frequency of intercourse, sexual
techniques. Absence of morning erections suggests an organic cause of
erectile dysfunction.
Physical examination
• Body hair distribution.
• Muscle mass and fat distribution.
• Eunuchoidism.
• Gynaecomastia.
• Genital examination:
• Pubic hair. Normal ♂ escutcheon.
• Phallus. Normal >5cm length and >3cm width.
• Testes. Size (using Prader orchidometer) and consistency (normal
>15m and firm).
• Look for nodules or areas of tenderness.
• General examination: look for evidence of systemic disease.
• Assess sense of smell and visual fields.
• Reflexes.
• Peripheral pulses.
• Rectal examination.
Gynaecomastia
Definition
Enlargement of the ♂ breast, as a result of hyperplasia of the glandular
tissue, to a diameter of >2cm (see Fig. 4.8). Common; present in up to
one-third of ♂ <30 years and in up to 50% of ♂ >45 years.
See Box 4.20 for causes.
Aromatase activity
(converts testosterone to LH/hCG
oestrogen)
SHBG
Testosterone Oestrogen (binds testosterone more
than oestrogen)
Evaluation of gynaecomastia
History
• Duration and progression of gynaecomastia.
• Further investigation warranted if:
• Rapidly enlarging gynaecomastia.
• Recent-onset gynaecomastia in a lean post-pubertal ♂.
• Painful gynaecomastia.
• Exclude underlying tumour, e.g. testicular cancer.
• Symptoms of hypogonadism. Reduced libido, erectile dysfunction.
• Symptoms of systemic disease, e.g. hepatic, renal, and endocrine disease.
• Drug history, including recreational drugs, e.g. alcohol.
Physical examination
• Breasts:
• Pinch breast tissue between thumb and forefinger—distinguish
from fat.
• Measure glandular tissue diameter. Gynaecomastia if >2cm.
• If >5cm, hard, or irregular, investigate further to exclude breast
cancer.
• Look for galactorrhoea.
• Testicular palpation:
• Exclude tumour.
• Assess testicular size—? atrophy.
• 2° sex characteristics.
• Look for evidence of systemic disease, e.g. chronic liver or renal
disease, thyrotoxicosis, Cushing’s syndrome, chronic cardiac or
pulmonary disease.
Investigations
(See Fig. 4.9.)
Baseline investigations
• Serum testosterone.
• Serum oestradiol.
• LH and FSH.
• Prolactin.
• SHBG.
• hCG.
• Liver function tests.
Additional investigations
• If testicular tumour is suspected, e.g. raised oestradiol/hCG:
testicular US.
• If adrenal tumour is suspected, e.g. markedly raised oestradiol:
dehydroepiandrosterone sulphate; abdominal CT or MRI scan.
• If breast malignancy is suspected: mammography; FNAC/tissue biopsy.
• If lung cancer is suspected, e.g. raised hCG: chest radiograph.
• Other investigations, depending on clinical suspicion, e.g. renal or
thyroid function.
EVALUATION OF GYNAECOMASTIA 385
Abdominal/
chest imaging
Extragonadal E2 or hCG
secreting tumour
Management of gynaecomastia
• Treat underlying disorder when present. Withdraw offending drugs
where possible.
• Reassurance in the majority of idiopathic cases. Often resolves
spontaneously.
• Treatment may be required for cosmetic reasons or to alleviate pain
and tenderness.
• Drug treatment only partially effective. May be of benefit in treating
gynaecomastia of recent onset.
Medical
See Table 4.17.
Surgical
Reduction mammoplasty may be required in ♂ with severe and persistent
gynaecomastia.
Further reading
Carlson HE (2011). Approach to the patient with gynecomastia. J Clin Endocrinol Metab 96, 15-21.
Khan HN, Blarney RW (2003). Endocrine treatment of physiological gynaecomastia. BMJ 327, 301–2.
TESTICULAR TUMOURS 387
Testicular tumours
Epidemiology
• 6/100,000 ♂ per year.
• Incidence rising, particularly in North West Europe.
See Table 4.18 for classification.
Risk factors
• Cryptorchidism.
• Gonadal dysgenesis.
• Infertility/reduced spermatogenesis.
Prognosis
Seminomas
• 95% cure for early disease; 80% cure for stages II/IV.
• i incidence of second tumours and leukaemias 20 years after therapy.
Non-seminoma germ cell tumours
• 90% cure in early disease, falling to 60% in metastatic disease.
• i incidence of second tumours and leukaemias 20 years after therapy.
Stromal tumours
• Excellent prognosis for benign tumours.
• Malignant tumours are aggressive and are poorly responsive to
treatment.
Further reading
Griffin JE, Wilson JD (1998). Disorders of the testes and male reproductive tract. In: Wilson JD,
Foster DW, Kronenberg HM, Larson PR (eds.) Williams textbook of endocrinology, 9th edn,
pp. 819–76. WB Saunders, Philadelphia.
388 CHAPTER 4 Reproductive endocrinology
Erectile dysfunction
Definition
The consistent inability to achieve or maintain an erect penis sufficient for
satisfactory sexual intercourse. Affects approximately 10% of ♂ and >50%
of ♂ >70 years.
Physiology of male sexual function
• The erectile response is the result of the coordinated interaction of
nerves, smooth muscle of the corpora cavernosa, pelvic muscles, and
blood vessels.
• It is initiated by psychogenic stimuli from the brain or physical
stimulation of the genitalia, which are modulated in the limbic
system, transmitted down the spinal cord to the sympathetic and
parasympathetic outflows of the penile tissue.
• Penile erectile tissue consists of paired corpora cavernosa on
the dorsum of the penis and the corpus spongiosum. These are
surrounded by fibrous tissue known as the tunica albuginea.
• In the flaccid state, the corporeal smooth muscle is contracted,
minimizing corporeal blood flow and enhancing venous drainage.
• Activation of the erectile pathway results in penile smooth muscle
relaxation and cavernosal arterial vasodilatation. As the corporeal
sinuses fill with blood, the draining venules are compressed against the
tunica albuginea, so venous outflow is impaired. This results in penile
rigidity and an erection.
• Corporeal vasodilatation is mediated by parasympathetic neuronal
activation, which induces nitric oxide release by the cavernosal nerves.
This activates guanyl cyclase, thereby i cGMP and causing smooth
muscle relaxation.
• Detumescence occurs after the inactivation of cGMP by the enzyme
phosphodiesterase, resulting in smooth muscle contraction and
vasoconstriction.
• Ejaculation is mediated by the sympathetic nervous system.
Pathophysiology
Erectile dysfunction may thus occur as a result of several mechanisms:
• Neurological damage.
• Arterial insufficiency.
• Venous incompetence.
• Androgen deficiency.
• Penile abnormalities.
ERECTILE DYSFUNCTION 389
390 CHAPTER 4 Reproductive endocrinology
Penile prosthesis
• Is usually tried in ♂ either reluctant to try other forms of therapy
or when other treatments have failed. They may be semi-rigid or
inflatable.
• Complications. Infection, mechanical failure.
Psychosexual counselling
Particularly for ♂ with psychogenic impotence and in ♂ who fail to
improve with the above therapies.
Surgical
• Rarely indicated, as results are generally disappointing.
• Revascularization techniques may be available in specialist centres.
• Ligation of dorsal veins may restore erectile function temporarily in
men with venous insufficiency, although rarely permanently.
Further reading
Beckman TJ, et al. (2006). Evaluation and medical management of erectile dysfunction. Mayo Clinic
Proc 81, 385–90.
Cohan P, Korenman SG (2001). Erectile dysfunction. J Clin Endocrinol Metab 86, 2391–4.
Fazio L, Brick G (2004). Erectile dysfunction: management update. Can Med Assoc J 170, 1429–37.
Shamloul R, Ghanem H (2013). Erectile dysfunction. Lancet 381, 153–65.
MANAGEMENT OF ERECTILE DYSFUNCTION 395
396 CHAPTER 4 Reproductive endocrinology
Infertility
Definition
• Infertility, defined as failure of pregnancy after 1 year of unprotected
regular (2x week) sexual intercourse, affects 710% of all couples.
• Couples who fail to conceive after 1 year of regular unprotected
sexual intercourse should be investigated.
• If there is a known predisposing factor or the ♀ partner is over
35 years of age, then investigation should be offered earlier.
Causes
(See Boxes 4.23 and 4.24.)
• ♀ factors (e.g. PCOS, tubal damage) 35%.
• ♂ factors (idiopathic gonadal failure in 60%) 25%.
• Combined factors 25%.
• Unexplained infertility 15%.
Investigations
(See Fig. 4.10.)
• General assessment:
• Measure TSH, free T4, and serum prolactin in ♀ with irregular
menstrual cycles or who are not ovulating.
• Serum testosterone, SHBG, 17OH progesterone if there is clinical
evidence of hyperandrogenism, in the presence of irregular
menstrual cycles, or if anovulation is confirmed.
• MRI of the pituitary fossa in hyperprolactinaemia and
hypogonadotrophic hypogonadism.
• Assess ovarian reserve:
• Check serum FSH and LH. In ♀ who are not amenorrhoeic, this
should be measured on days 2–6 of the menstrual cycle (day
1 = first day of menses). Follicular phase (FSH >10) is indicative of
reduced ovarian reserve. Ovarian failure is diagnosed if FSH >30.
• Pelvic US. Not only for uterine and ovarian anatomy but also for
antral follicle count (number of small follicles as an estimate of
ovarian ageing).
• Consider AMH measurement.
• Assess ovulatory function:
• Home ovulation kit testing of urinary LH reliable in regular cycles.
Home temperature testing less reliable.
• In ♀ with regular menstrual cycles—measure a midluteal
progesterone (day 21 or approximately 7 days before expected
onset of menses >30pmol/L consistent with ovulation).
• Consider ovulation tracking by serial ultrasound if uncertain.
• Exclude infection. Send vaginal discharge for bacteriology, and do
Chlamydia trachomatis serology.
• Assess tubal patency (refer to specialist multidisciplinary fertility clinic):
• Hysterosalpingography (HSG) or laparoscopy and dye test (do in
the early follicular phase of cycle to avoid doing during pregnancy).
400 CHAPTER 4 Reproductive endocrinology
Assess ovulation
in 2−3 cycles
If normal
semen analysis FSH, LH, E2
Laparoscopy
i FSH (>30) Normal FSH d FSH
Hypogonadotrophic
Tubal factor Normal POF PCOS hypogonadism
Semen analysis
Normal Abnormal
Prolactin
Karyotype
Pituitary MRI
Vaginal/cervical factors
• Each episode of acute PID causes infertility in 10–15% of cases.
• Trachomatis is responsible for half the cases of PID in developed
countries. Treat both partners with antibiotics.
Uterine factors
• Intracavity fibroids, polyps, uterine septum can be resected
hysteroscopically, with high chance of restoring fertility.
• Intramural fibroids may also reduce fertility and may require
myomectomy.
• Fibroid embolization is not recommended for ♀ wishing fertility, as
safety in pregnancy not established.
Table 4.21 Assisted reproductive techniques (ART)
Technique Indications Pregnancy rates Notes
Intrauterine insemination Unexplained infertility <15% per cycle Washed and prepared motile spermatozoa are inje
(IUI) (usually offered up Mild oligozoospermia 15% the uterine cavity through a catheter just before ov
to six cycles) (>2 x 106 motile sperm) Superovulation may improve success rates but is
Mild endometriosis associated with an increased risk of multiple pre
In vitro fertilization (IVF) Most forms of infertility 20–30% pregnancy rate per cycle After superovulation, ovarian follicles are aspirate
unless severe male factor 80–90% delivery rate after six ultrasonic guidance and are fertilized with prepare
cycles in women under the age in vitro. The embryos are then transferred back in
of 35 years uterine cavity, usually 48h after insemination. Lute
Success rates markedly reduced using progesterone supplementation (pessaries o
after 40 years of age then provided until pregnancy is confirmed.
Babies conceived by IVF have a 2x May adopt a similar technique in women with pr
increased risk of low birthweight ovarian failure using donated ova which are then
and preterm delivery in vitro with partner’s sperm. Hormonal support
required following embryo transfer.
Gamete intrafallopian Most forms of infertility Similar to IVF Similar to IVF, except that retrieved follicles and
transfer (GIFT) unless severe male factor injected laparoscopically into a Fallopian tube to
Do not use in women naturally.
with tubal disease Rarely performed in the UK, as it offers little adv
over IVF.
Intracytoplasmic sperm Male infertility 20–30% per cycle if female partner Viable spermatozoa are injected directly into ooc
injection (ICSI) under 40 years of age retrieved following superovulation. Embryos are
There is a small risk of sex then implanted into the uterus. Spermatozoa may
chromosome abnormalities in males concentrated from an ejaculate or be aspirated fr
(1%) conceived following ICSI epididymis or testis in men with obstructive azoo
408 CHAPTER 4 Reproductive endocrinology
Unexplained infertility
Definition
Infertility despite normal sexual intercourse occurring at least twice
weekly, normal semen analysis, documentation of ovulation in several
cycles, and normal patent tubes (by laparoscopy).
Management
30–50% will become pregnant within 3 years of expectant management. If
not pregnant by then, chances that spontaneous pregnancy will occur are
greatly reduced, and ART should be considered. In ♀ >34 years of age,
then expectant management is not an option, and up to six cycles of IUI
or IVF should be considered.
Results
• Superovulation with IUI can achieve a pregnancy rate of 15% per cycle
and cumulative delivery rate after several cycles of 50%.
• IVF offers a live birth rate per cycle of >30% in younger ♀ and a
cumulative delivery rate approaching 80%.
• ICSI allows IVF to be offered in severe oligospermia, with pregnancy
rates equivalent to standard IVF. Slight increase in congenital
malformations after ICSI. Significant risk of multiple pregnancies with
any form of ART.
410 CHAPTER 4 Reproductive endocrinology
Ovulation induction
Indications
• Anovulation due to:
• PCOS.
• Hypopituitarism.
• Hypogonadotrophic hypogonadism.
• Controlled ovarian hyperstimulation for IVF.
Pretreatment assessment
• Exclude thyroid dysfunction and hyperprolactinaemia.
• Check rubella serology.
• Confirm normal semen analysis.
• Confirm tubal patency (laparoscopy, hysterosalpingogram (HSG),
or hysterosalpingo-contrast-sonography (HyCoSy)) prior to
gonadotrophin use and/or after failed clomiphene use.
• Optimize lifestyle: maintain satisfactory BMI, exercise in moderation,
reduce alcohol intake, and stop smoking.
• Baseline pelvic US is essential to exclude ovarian masses and uterine
abnormalities prior to treatment.
Clomifene citrate
• Mode of action:
• Antioestrogen blocking normal –ve feedback, thereby i pulse
frequency of GnRH. This stimulates FSH and LH release, thereby
stimulating the production of one or more dominant ovarian follicles.
• Antioestrogen effect on endometrium, cervix, and vagina.
• Indications. Eugonadotrophic anovulation, e.g. PCOS. May also be used
in unexplained infertility. Requires normal hypothalamo–pituitary–
ovarian axis to work, therefore ineffective in hypogonadotrophic
hypogonadism.
• Administration. Start on days 2–5 of menstrual cycle (may have to
induce bleed by giving a progestagen for 10 days), and take for a total
of 5 days.
• Dose:
• Most require 50–100mg/day for 5 days.
• Should be used with ovulation tracking by ultrasound to avoid
multiple pregnancies and to confirm effectiveness.
• Spontaneous ovulation should occur 5–10 days after last day of
medication.
• Remain on optimum dose for 6–12 months.
• Efficacy:
• 80–90% ovulate, with conception rates of 50–60% in first six ovulatory
cycles. May enhance chances of ovulation in non-responders by
the administration of 10,000IU of hCG midcycle (use US guidance;
administer hCG when leading follicle is at least 20mm).
• Side effects:
• Hot flushes in 10%, mood swings, depression and headaches in 1%,
pelvic pain in 5%, nausea in 2%, breast tenderness in 5%, hair loss in
0.3%, visual disturbances in 1.5%.
OVULATION INDUCTION 411
Further reading
Braude P, Muhammed S (2003). Assisted conception and the law in the United Kingdom. BMJ
327, 978–81.
Farhat R, et al. (2010). Outcome of gonadotropin therapy for male infertility due to hypogonado-
trophic hypogonadism. Pituitary 13, 105–10.
Hamilton-Fairley D, Taylor A (2003). Anovulation. BMJ 327, 546–9.
Hirsh A (2003). Male subfertility. BMJ 327, 669–72.
Royal College of Obstetrics and Gynaecology (2004). Fertility assessment and treatment for peo-
ple with fertility problems. RCOG, pp.1–208. Royal College of Obstetrics and Gynaecology,
London.
414 CHAPTER 4 Reproductive endocrinology
Evaluation
• Karyotype. 46,XX vs 46,XY ♂ or ♀.
• Imaging:
• Look for presence of testes or ovaries and uterus.
• Most easily performed using pelvic US, but MRI may be more
sensitive in identifying internal genitalia and abnormally sited gonads
in cryptorchidism.
Hormonal evaluation
• LH, FSH, testosterone, oestradiol, SHBG.
• hCG stimulation test (b see Clinical assessment, p. 119)—to assess
the presence of functioning testicular material. Measure testosterone,
androstenedione, DHT, and SHBG post-stimulation.
• Others, depending on clinical suspicion, for example:
• 5A-reductase deficiency—check DHT levels before and after hCG
stimulation.
• 21-hydroxylase deficiency—17-hydroxyprogesterone 9 ACTH
stimulation (b see Investigations, p. 322).
• Urinary steroid profile—mass spectrometry and gas
chromatography as screen for some enzyme defects.
See Box 4.25 for causes.
DISORDERS OF SEXUAL DIFFERENTIATION 415
Ovotesticular DSD
Pathogenesis
• Unknown. May be familial.
• Previously known as true hermaphroditism.
• Affected individuals have both ovarian and testicular tissue, either in
the same gonad (ovotestis) or an ovary on one side and a testis on the
other. A uterus and a Fallopian tube are usually present, the latter on
the side of the ovary or ovotestis. Wolffian structures may be present
in a third of individuals on the side of the testis. The testicular tissue is
usually dysgenetic, although the ovarian tissue may be normal.
Clinical features
• Most individuals have ambiguous genitalia and are raised as ♂, but just
under 10% have normal ♀ external genitalia.
• At puberty, 50% of individuals menstruate, which may present as cyclic
haematuria in ♂, and most develop breasts.
• Feminization and virilization vary widely. Most are infertile, but fertility
has been reported.
• 2% risk of gonadal malignancy, higher in 46,XY individuals.
Evaluation
• 46,XX in 70%, 46,XX/46,XY in 20%, and 46,XY in 10%.
• Hypergonadotrophic hypogonadism is usual.
• Diagnosis can only be made on gonadal biopsy.
418 CHAPTER 4 Reproductive endocrinology
Transsexualism
Definition
A condition in which an apparently anatomically and genetically normal
person feels that he or she is a member of the opposite sex. Gender dys-
phoria is an irreversible discomfort with the anatomical gender, which may
be severe, often developing in childhood.
Epidemiology
• More common in ♂.
• Estimated prevalence of 1:13,000 ♂ and 1:30,000 ♀.
Aetiology
• Unknown and controversial.
• Some evidence that it may have a neurobiological basis. There appear
to be sex differences in the size and shape of certain nuclei in the
hypothalamus. ♂ to ♀ transsexuals have been found to have ♀
differentiation of one of these nuclei, whereas ♀ to ♂ transsexuals
have been found to have a ♂ pattern of differentiation.
Management
Standards of care
• Multidisciplinary approach between psychiatrists, endocrinologists, and
surgeons. Patient should be counselled about the treatment options,
risks, and implications, and realistic expectations should be discussed.
• Endocrine disorders should be excluded prior to entry into the gender
reassignment programme, i.e. ensure normal internal and external
genitalia, karyotype, gonadotrophins, and testosterone/oestradiol.
• Psychiatric assessment and follow-up is essential before definitive
therapy. The transsexual identity should be shown to have been
persistently present for at least 2 years to ensure a permanent diagnosis.
• Following this period, the subject should dress and live as a member
of the desired sex under the supervision of a psychiatrist. This should
continue for at least 3 months before hormonal treatment and 1 year
before surgery.
• Psychological follow-up and expert counselling should be available, if
required, throughout the programme, including after surgery.
• In adolescents, puberty can be reversibly halted by GnRH analogue
therapy to prevent irreversible changes in the wrong gender until a
permanent diagnosis is made.
• See Box 4.27 for monitoring of therapy.
TRANSSEXUALISM 421
Hormonal manipulation
See Box 4.26 for contraindications.
Male to female transsexuals
• Suppress ♂ s sex characteristics: cyproterone acetate (CPA)
(100mg/day).
• Induce ♀ s sex characteristics:
• Ethinylestradiol 100 micrograms/day or estradiol valerate 2mg bd/
tds or estradiol patch 100 micrograms 2x/week (stop smoking).
• Medroxyprogesterone acetate 10mg od.
• Aims:
• Breast development—maximum after 2 years of treatment.
• Development of ♀ fat distribution.
• d body hair and smoother skin. However, facial hair is often
resistant to treatment.
• d muscle bulk and strength.
• Reduction in testicular size.
• Hormonal manipulation has little effect on voice.
422 CHAPTER 4 Reproductive endocrinology
Endocrinology in
pregnancy
Maternal hyperthyroidism
(b see Thyrotoxicosis in pregnancy, p. 44.)
Incidence
• Affects 0.2% of pregnant women.
• Most are diagnosed before pregnancy or in the first trimester of
pregnancy.
• In women with Graves’s disease in remission, exacerbation may occur
in first trimester of pregnancy.
Graves’s disease
• The commonest scenario is pregnancy in a patient with pre-existing
Graves’s disease on treatment, as fertility is low in patients with
untreated thyrotoxicosis. Newly diagnosed Graves’s disease in
pregnancy is unusual.
• Aggravation of disease in first trimester, with amelioration in second
half of pregnancy because of a decrease in maternal immunological
activity at that time.
• Symptoms of thyrotoxicosis are difficult to differentiate from
normal pregnancy. The most sensitive symptoms are weight loss and
tachycardia. Goitre is found in most patients.
Management
• Risks of uncontrolled hyperthyroidism to mother: heart failure/
arrhythmias.
• Antithyroid drugs (ATDs) are the treatment of choice but cross the
placenta.
• Propylthiouracil (PTU) should be used in the first trimester in case
of teratogenic effects of carbimazole (check liver function monthly).
Thereafter, carbimazole is recommended.
• Carbimazole use in pregnancy was discouraged, as it was thought to be
associated with aplasia cutis, a rare scalp defect, though recent studies
have shown no increase in rate. Studies have shown that carbimazole
may be associated with choanal and oesophageal atresia, but the
maternal hyperthyroidism may be a factor in this.
• Propylthiouracil may rarely be associated with hepatocellular
inflammation, which, in severe cases, can lead to liver failure and
death. A recent study also showed that PTU was associated with low
birthweight infants.
• A short course of 40mg of propranolol tds can be used initially for 2–3
weeks while antithyroid drugs take affect.
• Most patients will be on a maintenance dose of ATD. A high dose of
ATD may be necessary initially to achieve euthyroidism as quickly as
possible (carbimazole 20–40mg/day or propylthiouracil 200–400mg/
day) in newly diagnosed patients, then use the minimal dose of ATD to
maintain euthyroidism.
• Do not use block and replace regime as higher doses of ATDs
required, and there is minimal transplacental transfer of T4, thereby
risking fetal hypothyroidism.
• Monitor TFTs every 4–6 weeks.
MATERNAL HYPERTHYROIDISM 429
• Aim to keep FT4 at upper limit of normal and TSH low normal
(<2.5mU/L—first trimester, <3.0mU/L—second trimester,
<3.5mU/L—third trimester).
• In 730% of women, ATD may be discontinued at 32–36 weeks’
gestation. Consider if euthyroid for at least 4 weeks on lowest dose
of ATD, but continue to monitor TFTs frequently. The presence of
a large goitre or ophthalmopathy suggests severe disease and the
chances of remission are low, so do not stop ATD. Graves’s disease
can flare in the post-natal period due to increase in the maternal
antibody levels, so patients should be monitored closely in the
post-partum period.
• Risks of neonatal hypothyroidism and goitre are reduced if woman on
carbimazole 20mg or less (200mg propylthiouracil or less) in last few
weeks of gestation.
• Breastfeeding is safe at doses of less than 150mg of PTU and 15mg
carbimazole, as only small amounts pass into the breast milk (<0.7%
PTU and 0.5% carbimazole).
• If higher doses are given, we recommend breastfeeding prior to taking
medication, and the dose can be divided throughout the day. Neonatal
thyroid function needs to be monitored.
• Surgical management of thyrotoxicosis is rarely necessary in pregnancy.
Only indication: serious ATD complication (e.g. agranulocytosis) or
drug resistance. There is a possible i risk of spontaneous abortion
or preterm delivery associated with surgery during pregnancy. The
second trimester is the optimal time for surgery, as organogenesis is
complete and less chance of anaesthesia and surgery inducing labour.
• Radioiodine therapy is contraindicated in pregnancy, during
breastfeeding, and for 4 months prior to conception.
Infants born to mothers with Graves’s disease
• Risks to fetus of uncontrolled thyrotoxicosis:
• i risk of spontaneous miscarriage and stillbirth.
• Intrauterine growth restriction (IUGR).
• Preterm labour.
• Fetal or neonatal hyperthyroidism.
• Follow-up of babies born to mothers on ATDs show normal weight,
height, and intellectual function.
• Fetal hypothyroidism. May occur following treatment of mother
with high doses of ATDs (>200mg PTU/day; >20mg carbimazole),
particularly in the latter half of pregnancy. This is rare and may be
diagnosed by demonstrating a large fetal goitre on fetal US in the
presence of fetal bradycardia.
• Fetal hyperthyroidism. May occur after week 25 of gestation. It results
in IUGR, fetal goitre, and tachycardia (fetal heart rate >160bpm) and
has high mortality if not treated. It may develop if the mother has
high titres of TSH-stimulating antibodies (TSAb) which can cross the
placenta. Treat by giving mother ATD, and monitor fetal heart rate (aim
<140bpm), growth, and goitre size. It is important to remember women
with Graves’s disease who have been treated with radioiodine or
surgery can still have high antibody titres and therefore need monitoring.
430 CHAPTER 5 Endocrinology in pregnancy
Maternal hypothyroidism
Prevalence
• 2.5% subclinical hypothyroidism.
• 1–2% overt hypothyroidism.
Risks of suboptimal treatment during pregnancy
• Spontaneous miscarriage. Twofold i risk.
• Pre-eclampsia. 21% of suboptimally treated mothers have
pregnancy-induced hypertension (PIH).
• Also i risk of anaemia during pregnancy and post-partum
haemorrhage.
• The fetus is dependent on maternal thyroxine until fetal production
starts at 12 weeks.
• Risk of impaired fetal intellectual and cognitive development.
• i risk of perinatal death.
• Other risks to fetus are those associated with PIH (IUGR, preterm
delivery, etc.).
• The risk of congenital malformations is not thought to be i.
Management
• Spontaneous pregnancy in overtly hypothyroid women is unusual, as
hypothyroid women are likely to have anovulatory menstrual cycles.
• Levothyroxine therapy. Start on 100 micrograms. Measure TSH and free
T4 4 weeks later.
• For women on thyroxine, optimize therapy prior to pregnancy.
• If already on T4 before pregnancy, on confirmation of pregnancy,
increase dose by 30% (25–50 micrograms) because of increased TBG
and thyroid hormone requirements.
• Aim. TSH—lower part of normal range; FT4—upper end of normal.
• Many women are suboptimally replaced prior to pregnancy, so their
dose needs to be increased in response to abnormal TFTs. After
delivery, they should continue on this increased dose, with monitoring
of biochemistry.
• NB Do not give FeSO4 simultaneously with T4—reduces its efficacy.
Separate times for drug ingestion by at least 2h.
Causes of maternal hypothyroidism
• Hashimoto’s thyroiditis (most common cause).
• Previous radioiodine therapy or thyroidectomy.
• Previous post-partum thyroiditis.
• Hypopituitarism.
Positive thyroid antibodies but euthyroid
• Twofold excess risk of spontaneous miscarriage.
• No other complications.
• No risk of neonatal hypothyroidism.
• Risk of PIH not i.
• The occasional mother will develop hypothyroidism towards the end
of the pregnancy, so check TFTs between weeks 28–32 of gestation.
• i risk of post-partum thyroiditis, so check TSH at 3 months
post-partum.
432 CHAPTER 5 Endocrinology in pregnancy
Parathyroid disorders
Calcium metabolism in pregnancy
• There are increased calcium requirements in pregnancy; 25–30g is
transferred to the baby.
• There is increased maternal intestinal absorption of calcium.
• Due to increased renal blood flow, there is increase in the renal
excretion of calcium.
Primary hyperparathyroidism in pregnancy
• Rare—8 cases in 100,000 in women of childbearing age.
• In pregnancy, the hypercalcaemia may improve due to the fetal transfer
of calcium.
• Diagnosis is made by finding a raised PTH in the presence of a raised
calcium.
• Ultrasound can be used to locate the adenoma.
• Can cause serious maternal morbidity which includes hyperemesis,
nephrolithiasis, peptic ulcers, and pancreatitis. It is also associated with
pre-eclampsia, recurrent miscarriage, preterm labour.
• Fetal complications include IUGR and neonatal death. The high
circulating maternal calcium can lead to suppression of fetal
parathyroid development, leading to hypocalcaemia of the fetus after
delivery. Neonates normally present at days 5–14 with poor feeding,
tetany, and convulsions.
Management
• Hypercalcaemia is treated with IV fluids.
• If mild, women can be advised to increase their fluid intake. Calcium
levels should be monitored throughout pregnancy, with surgery
arranged post-partum.
• If calcium remains persistently high (>2.85mmol/L), then
parathyroidectomy can be arranged in the second or early third
trimester.
Hypoparathyroidism
• Pregnancy increases the demand for vitamin D. Therefore,
replacement doses will need to be increased in pregnancy.
• Maternal levels of corrected calcium and vitamin D should be checked
monthly.
• Untreated hypoparathyroidism leads to miscarriage, fetal
hypocalcaemia, and neonatal rickets.
PARATHYROID DISORDERS 435
Further reading
Abalovich M, Amino N, Barbour LA, et al. (2007). Management of thyroid dysfunction during preg-
nancy and post-partum: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab
92(8 Suppl), S1–47.
Alexander EK, Marqusee E, Lawrence J, et al. (2004). Timing and magnitude of increases in levothy-
roxine requirements during pregnancy in women with hypothyroidism. N Engl J Med 351, 241–9.
Azizi F, Amouzegar A (2011). Management of hyperthyroidism during pregnancy. Eur J Endocrinol
164, 871–6.
Chen C, Xirasager S, et al. (2011). Risk of adverse perinatal outcomes with antithyroid treatement
during pregnancy: a national population based study. BJOG 118, 1365–73.
Cooper S, Rivkees S (2009). Putting propylthiouracil in perspective. J Clin Endocrinol Metab 94,
1881–2.
De Groot L, et al. (2012). Management of thyroid dysfunction during pregnancy and post-partum: an
Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 97, 2543–65.
Lazarus JH. (2011).Thyroid function in pregnancy. Br Med Bull 97, 137–48.
LeBeau SO, Mandell SJ (2006). Thyroid disorders during pregnancy. Endocrinol Metab Clin North
Am 35, 117–36.
Norman J, et al. (2009). Hyperparathyroidism during pregnancy and the effect of rising calcium on
pregnancy loss: a call for earlier intervention. Clin Endocrinol (Oxf) 71, 104–9.
Poppe K, Glinoer D (2003). Thyroid autoimmunity and hypothyroidism before and during preg-
nancy. Hum Reprod Update 9, 149–61.
Sato K (2008). Hypercalcemia during pregnancy, puerperium and lactation: Review and a case
report of hypercalcemic crisis after delivery due to excessive productin of PTH-related protein
(PTHrP) without malignancy (humoral hypercalcemia of pregnancy). Endocrin J 55, 959–66.
Stagnaro-Green A (2002). Post-partum thyroiditis. J Clin Endocrinol Metab 87, 4042–7.
436 CHAPTER 5 Endocrinology in pregnancy
Pituitary disorders
Normal anatomical changes during pregnancy
• Prolactin (PRL)-secreting cells. Marked lactotroph hyperplasia during
pregnancy.
• Gonadotrophin-secreting cells. Marked reduction in size and number.
• TSH and ACTH-secreting cells. No change in size or number.
• Anterior pituitary. Size increases by up to 70% during pregnancy. May
take 1 year to shrink to near pre-pregnancy size in non-lactating
women. Gradual slight increase in size with each pregnancy.
• MRI. Enlarged anterior pituitary gland, but stalk is midline. Posterior
pituitary gland may not be seen in late pregnancy.
Normal physiology during pregnancy
• Serum PRL. Concentrations increase markedly during pregnancy and fall
again to pre-pregnancy levels approximately 2 weeks post-partum in
non-lactating women.
• Serum LH and FSH. Undetectable levels in pregnancy and blunted
response to GnRH because of –ve feedback inhibition from high levels
of sex hormones and PRL.
• Serum TSH, T4, and T3. TSH may be suppressed in the first trimester
of pregnancy. Free thyroid hormones usually at the lower end of the
normal range.
• Growth hormone (GH) and IGF-I. Low maternal GH levels and blunted
response to hypoglycaemia due to placental production of GH-like
substance. IGF-I levels are normal or high in pregnancy.
• ACTH and cortisol. CRH, ACTH, and cortisol levels are high in
pregnancy, as both CRH and ACTH are produced by the placenta. In
addition, oestrogen-induced increase in cortisol-binding globulin (CBG)
synthesis during pregnancy will further increase maternal plasma
cortisol concentrations. During the latter half of pregnancy, there is
a progressive increase of ACTH and cortisol levels, peaking during
labour. Incomplete suppression of cortisol, following dexamethasone
suppression test, and exaggerated response of cortisol to CRH
stimulation. However, normal diurnal variation persists.
PROLACTINOMA IN PREGNANCY 437
Prolactinoma in pregnancy
Effect of pregnancy on tumour size
Risk of significant tumour enlargement (i.e. resulting in visual field distur-
bances or headaches):
• Microadenoma 1–2%.
• Macroadenoma 15–35%.
• Macroadenoma treated with surgery and/or radiotherapy before
pregnancy 4–7%.
Effect of dopamine agonists on the fetus
Bromocriptine
Over 6,000 pregnancies have occurred in women receiving bromocriptine
in early pregnancy, and the incidence of complications in these pregnancies
with regard to fetal outcome is similar to that of the normal population,
indicating that bromocriptine is probably safe in early pregnancy. Data are
available on children whose mothers received bromocriptine throughout
pregnancy, and again the incidence of congenital abnormalities is negligible.
Children who are exposed to bromocriptine in utero have normal psycho-
logical development in follow-up.
The FDA has withdrawn the licence for use of bromocriptine
post-partum to suppress lactation, as there was an increased incidence
of adverse incidents which included myocardial infarction and stroke. We
would advise caution, particularly in women with pre-eclampsia.
Cabergoline
Also probably safe in early pregnancy and has been used in >380 pregnan-
cies, with no i risk of fetal loss or congenital abnormalities, but fewer data
are available. There are more data on the long-term safety of bromocrip-
tine, but cabergoline is being used increasingly as it is better tolerated.
Quinagolide
It has been used in >176 pregnancies, with a slightly higher rate of congeni-
tal abnormalities compared to the background rate.
Recent controversy
The Medicine and Health Regulatory Authority issued guidance on the
use of cabergoline in pregnancy, stating it should be stopped 1 month
pre-pregnancy. This recommendation is based on data in Parkinson’s
patients where high doses are used and an increased rate of cardiac and
pulmonary fibrosis has been reported. There are no published data on
similar conditions occurring in the fetus. Current practice is to use bro-
mocriptine first-line in women hoping to conceive, and cabergoline if they
cannot tolerate bromocriptine. Quinagolide is reserved for patients who
are unable to tolerate the other dopamine agonists.
438 CHAPTER 5 Endocrinology in pregnancy
Management of prolactinoma
Microprolactinoma
• After recent MRI, initiate dopamine agonist therapy to induce normal
ovulatory cycles and fertility.
• Stop bromocriptine as soon as pregnancy is confirmed.
• Assess for visual symptoms and headache at each trimester, although
the risk of complications is low (<5%). Serum PRL levels are difficult to
interpret during pregnancy, as they are normally elevated; therefore,
they are not measured.
• MRI is indicated in the occasional patient who becomes symptomatic.
• In the post-partum period, recheck serum PRL level 2 months after
cessation of breastfeeding. Reassess size of microprolactinoma by MRI
only if serum PRL level is higher than pre-pregnancy concentrations.
• 40–60% chance of remission of microprolactinoma following
pregnancy.
Macroprolactinoma
Management is controversial and must, therefore, be individualized. Three
possible approaches:
• Bromocriptine (because cabergoline does not have a licence in
pregnancy) may be used throughout pregnancy to reduce the risk
of tumour growth. The patient is monitored by visual fields at each
trimester or more frequently if symptoms of tumour enlargement
develop. This is probably the safest, and thus preferred, approach.
• May use bromocriptine or cabergoline to induce ovulation, and then
stop it after conception. However, patient must be monitored very
carefully during pregnancy with monthly visual field testing.
• If symptoms of tumour enlargement develop or there is deterioration
in visual fields, then MRI should be performed to assess tumour
growth. If significant tumour enlargement develops, then
bromocriptine therapy should be initiated.
• Alternatively, the patient may undergo surgical debulking of the
tumour and/or radiotherapy before seeking fertility. This will
significantly reduce the risk of complications associated with
tumour growth. However, this approach may render them
gonadotrophin-deficient. These patients should again be monitored
during pregnancy, using regular visual fields.
MRI should be performed in the post-partum period in women with
macroprolactinomas to look for tumour growth.
Breastfeeding
• There is no contraindication to breastfeeding.
• If the mother would like to breastfeed, dopamine receptor agonists
will have to be discontinued prior to birth, as they inhibit lactation.
The decision to discontinue medication should be assessed on a
case-by-case basis, dependent upon the potential risk of optic nerve/
chiasm compression.
CUSHING’S SYNDROME 439
Cushing’s syndrome
• Pregnancy is rare in women with untreated Cushing’s syndrome, as
75% of them will experience oligo- or amenorrhoea.
• Adrenal disease is the most common cause of Cushing’s syndrome
developing in pregnancy, responsible for over 50% of reported cases.
• The diagnosis of Cushing’s syndrome is difficult to establish during
pregnancy. However, the presence of purple striae and proximal
myopathy should alert the physician to the diagnosis of Cushing’s
syndrome.
• If suspected, the investigation of Cushing’s syndrome should be carried
out as in the non-pregnant state.
• The circulating levels of cortisol rise during normal pregnancy. This
is due to a combination of the increased levels of cortisol-binding
globulin, stimulated by the raised oestrogen, and the placental CRH
production which is biologically active.
• 24h urinary free cortisol measurements remain normal in the first
trimester but can increase by 3x by term in a normal pregnancy.
• Non-suppression of cortisol production on a low-dose dexamethasone
suppression test may be a feature of a normal pregnancy. There are no
internationally agreed normal values in pregnancy, so interpretation of
dynamic testing is difficult.
• The diurnal variation of cortisol secretion is, however, preserved in
normal pregnancy.
• Diagnosis is important, as pregnancy in Cushing’s syndrome is
associated with a high risk of maternal and fetal complications (see
Table 5.1).
440 CHAPTER 5 Endocrinology in pregnancy
Acromegaly
• Fertility in acromegaly is reduced, partly due to hyperprolactinaemia (if
present), in addition to secondary hypogonadism. However, there have
been several reported cases of pregnancy in acromegaly.
• Diagnosis in pregnancy is difficult, as the placenta secretes growth
hormone and, though different from maternal GH, assays may not be
able to detect this. IGF-1 also increases in normal pregnancy.
• Acromegaly increases the risk of gestational diabetes and hypertension,
so women should be screened.
• However, in the absence of diabetes mellitus, there does not appear
to be an excess of perinatal morbidity or mortality in babies born to
women with acromegaly.
• Significant tumour enlargement occasionally occurs, together with
enlargement of the normal pituitary lactotrophs, so monthly visual
field testing is recommended.
• Bromocriptine can be used in pregnancy, but there is less response
compared to prolactinomas.
• There are limited reports of somatostatin analogues and somatostatin
receptor agonists use in pregnancy, with no apparent adverse events.
They do cross the placenta. We would not advocate their use until
more data are available on their safety in pregnancy. Women are
generally advised to stop medication on conception.
• Headaches may be helped with both somatostatin analogues and
bromocriptine.
• Treatment may be deferred until after delivery in the majority of
patients.
Hypopituitarism in pregnancy
Pre-existing hypopituitarism
• Most commonly due to surgical treatment of and/or radiotherapy for a
pituitary adenoma.
• May require ovulation induction and thus conception by
gonadotrophin stimulation.
Lymphocytic hypophysitis
(b see also Lymphocytic hypophysitis, p. 194.)
• Rare disorder thought to be autoimmune in origin.
• Characterized by pituitary enlargement on imaging and variable loss of
pituitary function.
• Most commonly seen in women in late pregnancy or in the first year
post-partum.
• Symptoms are due to pressure effects, e.g. visual field defects and
headaches, or due to hormonal deficiency.
• Most common hormonal deficiencies:
• ACTH and vasopressin deficiency.
• TSH deficiency may also exist.
• Gonadotrophins and GH levels are usually normal.
• PRL levels may be mildly elevated in a third, and low in a third.
• Differential diagnosis:
• Pituitary adenoma.
• Sheehan’s syndrome.
• MRI often reveals diffuse homogeneous contrast enhancement of the
pituitary gland. However, the diagnosis is often only made definitively
by pituitary biopsy.
• There is an association with other autoimmune diseases, particularly
Hashimoto’s thyroiditis.
• Course variable. Pituitary function may deteriorate or improve with time.
Management
• Pituitary hormone replacement therapy, as required.
• Surgical decompression if pressure symptoms persist.
• A course of high-dose steroid therapy is controversial, with mixed
results.
Causes of hypopituitarism during pregnancy
• Pre-existing hypopituitarism.
• Pituitary adenoma.
• Lymphocytic hypophysitis.
• Sheehan’s syndrome.
Management of pre-existing hypopituitarism during pregnancy
• Hydrocortisone. Dose may need to be i in the third trimester of
pregnancy by 10mg a day, as the increase in CBG will reduce the
bioavailability of hydrocortisone. Parenteral hydrocortisone in a dose of
100mg IM every 6h should be given during labour and the dose reduced
back to maintenance levels in the post-partum period (24–72h).
HYPOPITUITARISM IN PREGNANCY 443
Adrenal disorders
Normal changes during pregnancy
Changes in maternal adrenocortical function
Markedly i concentrations of all adrenal steroids due to i synthesis and
d catabolism.
Feto-placental unit
• Fetal adrenal gland. DHEAS is produced in vast quantities by the fetal
adrenal gland. This is the major precursor for oestrogen synthesis
by the placenta. The fetal adrenal gland has a large capacity for
steroidogenesis. Stimulus for fetal adrenal gland unknown—possibly
hCG or PRL.
• Placenta:
• Maternal glucocorticoids are largely inactivated in the placenta by
11B-HSD. Maternal androgens are converted to oestrogens by
placental aromatase, thus protecting 5 fetus from virilization.
• Maternal catecholamines are broken down by placental
catechol-O-methyl transferase and monoamine oxidase activity.
ADDISON’S DISEASE IN PREGNANCY 445
Phaeochromocytoma
• Rare but potentially lethal in pregnancy. Maternal mortality may still
be as high as 17% and fetal mortality as high as 30% if not treated
promptly.
• Maternal death can be due to stroke, pulmonary oedema, arrhythmia,
and myocardial infarction, with the greatest risk during labour.
Placental vasoconstriction can lead to spontaneous fetal death,
intrauterine growth restriction, and fetal hypoxia.
• Hypertensive crisis can be precipitated by labour, delivery, opiates,
metoclopramide, and general anaesthesia.
• Women can present with palpitations, sweating, headache, anxiety,
dyspnoea, vomiting, and hyperglycaemia.
• Phaeochromocytoma needs to be differentiated from women with
pre-eclampsia, so suspect in women with hypertension, persistent or
intermittent, especially in:
• The absence of proteinuria or oedema, or presence of other
features.
• Hypertension developing before 20 weeks’ gestation, or
• Persistent glycosuria.
• Prenatal screening in high-risk women, e.g. those with a history or
family history of MEN-2 or von Hippel–Lindau syndrome.
• Diagnose by 24h urinary catecholamine collection (normal ranges are
unaltered in pregnancy) or raised plasma catecholamines.
• Labetalol and methyldopa can give false +ves.
• Tumour localization is important—MRI is the imaging of choice in
pregnancy.
448 CHAPTER 5 Endocrinology in pregnancy
Management of phaeochromocytoma
• A-blockade: phenoxybenzamine. Reduces fetal and maternal morbidity
and mortality. Appears to be safe in pregnancy. The starting dose is
10mg 12-hourly and is built up gradually to a maximum of 20mg every
8h. Oral prazosin and IV phentolamine can also be used.
• B-blockade: propranolol. Only after adequate A-blockade. May increase
the risk of intrauterine fetal growth restriction if started in the first
trimester, but benefits outweigh risks. Give in a dose of 40mg 8-hourly.
• Surgery. Timing is controversial. Some recommend, before 24 weeks’
gestation, surgical removal of phaeochromocytoma (relatively safe
following A- and B-blockade). After 24 weeks’ gestation, surgery
should be deferred until fetal maturity. The preferred method of
delivery is usually elective Caesarean section. Ensure adequate
adrenergic blockade before surgery. Surgery can be delayed until after
delivery.
Conn’s syndrome
• Rare in pregnancy, with <50 cases reported.
• Diagnosed with hypertension and hypokalaemia.
• Has been associated with placental abruption.
• Maternal mortality has been reported.
Diagnosis
• A suppressed renin and raised aldosterone (compared to normal
pregnancy ranges) confirm the diagnosis.
• US or MRI can be used to localize tumour.
Management
• Standard treatment for hypertension in pregnancy.
• Amiloride can be used in pregnancy.
• Spironolactone should be avoided, as associated with ambiguous
sexual genitalia in male rats.
Further reading
Ahlawat SK, Jain S, Kumaro S, Varma S, Sharma BK (1999). Phaeochromocytoma associated with
pregnancy: case report and review of the literature. Obstet Gynecol Surv 54, 728–37.
Hadden DR (1995). Adrenal disorders of pregnancy. Endocrinol Metab Clin North Am 24, 139–51.
Lindsay JR, Nieman LK (2006). Adrenal disorders in pregnancy. Endocrinol Metab Clin North Am
35, 1–20.
Robar C, Porremba J, Pelton J, Hudson L, Higby K (1998). Current diagnosis and management of
aldosterone-producing adenomas during pregnancy. Endocrinologist 8, 403–8.
Sam S, Molitch M (2003). Timing and special concerns regarding endocrine surgery during preg-
nancy. Endocrinol Metab Clin North Am 32, 337–54.
Chapter 6 449
Lining cells
Bone lining cells
d
eoi Formation
Osteoclasts ost
alized
mi ner
Osteocytes Un Mineralized
bone
Resorption
21 21 100
Days of remodelling cycle
120
100
(% female peak)
80
Bone mass
60
40
20
0
0 20 40 60 80
Age (years)
Fig. 6.2 Bone mass and age.
CALCIUM 451
Calcium
Roles of calcium
• Skeletal strength.
• Neuromuscular conduction.
• Stimulus secretion coupling (e.g. chromaffin cells).
Calcium in the circulation
Circulating calcium exists in several forms (see Fig. 6.3).
• Ionized—biologically active.
• Complexed to citrate, phosphate, etc.—biologically active.
• Bound to protein, mainly albumin—inactive.
Ionized
45% 50% Complexed
Protein bound
5%
Fig. 6.3 Forms of circulating calcium.
452 CHAPTER 6 Calcium and bone metabolism
Investigation of bone
Bone turnover markers
Used in some centres.
May be useful in:
• Assessing overall risk of osteoporotic fracture.
• Judging response to treatments for osteoporosis.
Resorption markers
• Collagen crosslinks. These are products of collagen degradation. The
small fragments of the ends of the collagen molecule are known as
telopeptides (NTX and CTX), and the measurement of these in blood
is the preferred biochemical measure of bone resorption.
Formation markers
• Total alkaline phosphatase is not specific to bone and is also found in
liver, intestine, and placenta. It is also insensitive to small changes in
bone turnover and is only of general use in monitoring the activity of
Paget’s disease.
• Bone-specific alkaline phosphatase is a more specific and reliable
measure of bone formation.
• Osteocalcin is a component of bone matrix, and the serum level of
osteocalcin reflects osteoblast activity.
• P1NP is a procollagen fragment released from the N terminal as type
1 collagen is laid down. When measured in serum, it is the most
sensitive and specific marker of bone formation.
The clinical utility of routine measurements of bone turnover markers is
not yet established.
BONE IMAGING 453
Bone imaging
Skeletal radiology
• Useful for:
• Diagnosis of fracture.
• Diagnosis of specific diseases (e.g. Paget’s disease and
osteomalacia).
• Identification of bone dysplasia.
• Not useful for assessing bone density.
Isotope bone scanning
Bone-seeking isotopes, particularly 99mtechnetium-labelled bisphospho-
nates, are concentrated in areas of localized i bone cell activity. Isotope
bone scans are useful for identifying localized areas of bone disease, such
as fracture, metastases, or Paget’s disease. However, isotope uptake is not
selective, and so i activity on a scan does not indicate the nature of the
underlying bone disease. Hence, subsequent radiology of affected regions
is needed to establish the diagnosis.
Isotope bone scans are particularly useful in Paget’s disease to establish
the extent and sites of skeletal involvement and the underlying disease
activity.
454 CHAPTER 6 Calcium and bone metabolism
Bone biopsy
Bone biopsy is occasionally necessary for the diagnosis of patients with
complex metabolic bone diseases. This is usually in the context of sus-
pected osteomalacia. Bone biopsy is not indicated for the routine diagno-
sis of osteoporosis. It should only be undertaken in highly specialist centres
with appropriate expertise.
456 CHAPTER 6 Calcium and bone metabolism
Urine excretion
Calcium
Measurement of 24h urinary excretion of calcium provides a measure
of risk of renal stone formation or nephrocalcinosis in states of chronic
hypercalcaemia. In other circumstances, particularly in the assessment
of the cause of hypercalcaemia (p hyperparathyroidism versus familial
hypocalciuric hypercalcaemia), an estimate of the renal handling of cal-
cium is more useful. This is most commonly estimated from the ratio of
the renal clearance of calcium to that of creatinine in the fasting state (see
Box 6.2). If all values are in mmol/L, the ratio is typically >0.02 in p hyper-
parathyroidism; values <0.01 are suggestive of hypocalciuric hypercalcae-
mia. (Exclude other causes of hypocalciuria, including renal insufficiency,
vitamin D deficiency.)
Phosphate
A 24h measurement of phosphate excretion largely reflects dietary phos-
phate intake and has little clinical utility.
Calcium-regulating hormones
Parathyroid hormone
Careful attention should be paid to local requirements for collecting blood
for PTH measurement. Since PTH secretion is suppressed by calcium
ingestion, it should be measured in the fasting state. The reference range
depends on the precise assay employed, but typical values are 10–60pg/
mL (1–6pmol/L).
Vitamin D and its metabolites
25OH vitamin D (25OHD)
This is the main storage form of vitamin D, and the measurement of ‘total
vitamin D’ is the most clinically useful measure of vitamin D status.
Internationally, there remains controversy around a ‘normal’ or ‘opti-
mal’ concentration of vitamin D. Levels over 50nmol/L are generally
accepted as satisfactory and values <25nmol/L representing deficiency.
True osteomalacia occurs with vitamin D values <15nmol/L.
Low levels of 25OHD can result from a variety of causes (b see
Vitamin D deficiency, p. 484). It is unlikely that serious intoxication will
occur with 25OHD concentrations of <125nmol/L.
1,25(OH)2 vitamin D (1,25(OH)2D)
Although this is the active form of vitamin D, measurement of its concen-
tration is rarely indicated. It is sometimes useful diagnostically in condi-
tions of extrarenal synthesis of 1,25(OH)2D, such as in sarcoidosis.
Parathyroid hormone-related peptide (PTHrP)
It is possible to measure the level of this oncofetoprotein in serum, but this
is very rarely indicated. Sample collection involves specific requirements.
Calcitonin
Calcitonin assays are available, but their utility is confined to the diagnosis
and monitoring of medullary carcinoma of the thyroid. There is no role
for calcitonin measurements in the routine investigation of calcium and
bone metabolism.
CALCIUM-REGULATING HORMONES 459
460 CHAPTER 6 Calcium and bone metabolism
Hypercalcaemia
Epidemiology
Hypercalcaemia is found in 5% of hospital patients and in 0.5% of the gen-
eral population.
Causes
Many different disease states can lead to hypercalcaemia. These are listed
by order of importance in hospital practice in Box 6.3. In asymptomatic
community-dwelling subjects, the vast majority of hypercalcaemia is the
result of hyperparathyroidism.
Clinical features
Notwithstanding the underlying cause of hypercalcaemia, the clinical fea-
tures are similar. With adjusted serum calcium levels <3.0mmol/L, signifi-
cant related symptoms are unlikely. With progressive increases in calcium
concentration, the likelihood of symptoms increases rapidly.
The clinical features of hypercalcaemia are well recognized (see Box
6.4); unfortunately, they are non-specific and may relate to underlying
illness.
Clinical signs of hypercalcaemia are rare. With the exception of band
keratopathy, these are not specific. It is important to seek clinical evidence
of underlying causes of hypercalcaemia, particularly malignant disease.
In addition to specific symptoms of hypercalcaemia, symptoms of
long-term consequences of hypercalcaemia should be sought. These
include the presence of bone pain or fracture and renal stones. These
indicate the presence of chronic hypercalcaemia.
Investigation of hypercalcaemia
Confirm the diagnosis
Serum calcium (adjusted for albumin).
Determine the mechanism
• i PTH. Parathyroid overactivity (p or tertiary hyperparathyroidism
can also occur in familial hypocalciuric hypercalcaemia and in lithium
therapy due to faulty calcium-sensing).
• d PTH. Parathyroid-independent cause.
• Normal PTH:
• May imply parathyroid overactivity—incomplete suppression.
• May imply altered calcium sensor—familial hypocalciuric
hypercalcaemia—calcium/creatinine excretion ratio will be low.
• Urine calcium to determine calcium/creatinine excretion ratio (not
correlated with the risk of stones).
Seek underlying illness (where indicated)
• History and examination.
• Chest X-ray.
• FBC and ESR.
• Biochemical profile (renal and liver function).
• Thyroid function tests (exclude thyrotoxicosis).
HYPERCALCAEMIA 461
Primary hyperparathyroidism
Present in up to 1 in 500 of the general population where it is predomi-
nantly a disease of post-menopausal ♀ (14/100,000 ♂, 28/100,000 ♀).
The normal physiological response to hypocalcaemia is an increase in
PTH secretion. This is termed s hyperparathyroidism and is not patho-
logical in as much as the PTH secretion remains under feedback control.
Continued stimulation of the parathyroid glands can lead to autonomous
production of PTH. This, in turn, causes hypercalcaemia which is termed
tertiary hyperparathyroidism. This is usually seen in the context of renal dis-
ease but can occur in any state of chronic hypocalcaemia, such as vitamin
D deficiency or malabsorption.
Pathology
• 85% single adenoma.
• 14% hyperplasia (may be associated with other endocrine
abnormalities, particularly multiple endocrine neoplasia (MEN) types
1 and 2 (b see MEN type 1, pp. 586–7; MEN type 2, pp. 592–3).
• <1% carcinoma (express the lectin galectin-3).
Clinical features
• Majority of patients are asymptomatic.
• Features of hypercalcaemia.
• End-organ damage—see Box 6.5.
Natural history
• In majority of patients without end-organ damage, disease is benign
and stable.
• A significant minority (2–3% per annum) will develop new indications
for surgery.
• Excess deaths probably linked to cardiovascular diseases and possibly
malignancy.
Investigation
(See Box 6.6.) Potential diagnostic pitfalls:
• FHH—differentiate with calcium/creatinine excretion ratio (b see
Familial hypocalciuric hypercalcaemia (FHH), pp. 457, 470) (<0.01 in
FHH; >0.02 in hyperparathyroidism).
• Long-standing vitamin D deficiency where the concomitant
osteomalacia and calcium malabsorption can mask hypercalcaemia
which becomes apparent only after vitamin D repletion. Consider
other causes of a raised PTH (see Box 6.7).
• Drugs associated with hypercalcaemia (e.g. thiazides and lithium).
Investigation is, therefore, primarily aimed at determining the presence of
end-organ damage from hypercalcaemia in order to determine whether
operative intervention is indicated.
PRIMARY HYPERPARATHYROIDISM 463
Treatment of hyperparathyroidism
Parathyroid surgery
(Aspects of parathyroid surgery also covered on b see p. 608.)
• For indications, see Table 6.2.
• Only by experienced surgeon (>20 procedures per year):
• Adenoma. Remove affected gland—often by minimally invasive
surgery.
• Hyperplasia. Partial parathyroidectomy (perhaps with reimplantation
of tissue in more accessible site).
Observation
• Suitable for patients with mild disease with no evidence of end-organ
damage.
• Most such patients have stable disease over many years but do require
monitoring:
• Annual serum calcium and renal function, BP.
• Every 2–3 years—BMD, renal US.
• Any significant deterioration is an indication for surgery.
Medical management
Only indicated if patient not suitable for surgery.
• Hormone replacement therapy:
• Preserves bone mass.
• Consider long-term risks (breast cancer, venous thrombosis, heart
disease, and stroke).
• Bisphosphonates:
• Clinically inconsequential effect on plasma and urine calcium.
• Preserve bone mass.
• Calcium-sensing receptor agonists:
• Cinacalcet (30mg twice daily) reduces serum but not urinary calcium
concentration. It increases sensitivity of calcium-sensing receptor,
decreasing PTH secretion. It is licensed for patients with severe
s hyperparathyroidism on dialysis, parathyroid carcinoma, and
patients with PHP in whom parathyroid surgery is contraindicated
or clinically inappropriate. It may also be used if the neck has been
explored and the parathyroid adenoma has not been found.
Indications for surgery in primary hyperparathyroidism
It is generally accepted that all patients with symptomatic hyperparathy-
roidism or evidence of end-organ damage should be considered for par-
athyroidectomy. This would include:
• Definite symptoms of hypercalcaemia. There is less good evidence
that non-specific symptoms, such as abdominal pain, tiredness, or mild
cognitive impairment, benefit from surgery.
• Impaired renal function.
• Renal stones (symptomatic or on radiograph).
• Parathyroid bone disease, especially osteitis fibrosis cystica.
• Pancreatitis.
TREATMENT OF HYPERPARATHYROIDISM 467
References
1. Bilezikian JP, Khan AA, Potts JT Jr, et al. (2009). Guidelines for the management of asymptomatic
primary hyperparathyroidism: Summary Statement from the Third International Workshop. J
Clin Endocrinol Metab 94, 335–9.
2. Davies M, Fraser WD, Hoskin DJ (2002). The management of primary hyperparathyroidism. Clin
Endocrinol (Oxf) 57, 145–55.
468 CHAPTER 6 Calcium and bone metabolism
Complications of parathyroidectomy
Mechanical
• Vocal cord paresis:
• May be permanent, particularly with repeated surgery.
• Tracheal compression from haematoma.
Metabolic (hypocalcaemia)
• Transient:
• Due to suppression of remaining glands.
• May sometimes require oral therapy with calcium 9 vitamin D
metabolites.
• Severe:
• Due to hungry bones—rare. Treatment with calcitriol 1 microgram/
day (sometimes higher dose required) and oral calcium, 3× daily,
may be required for several weeks.
• Occurs in patients with pre-existing bone disease.
• Minimize risk by pretreatment of any vitamin D deficiency for
several weeks. Risk of significant worsening of hypercalcaemia is
low, but monitoring of serum calcium is recommended.
• Acute severe hypocalcaemia requires initial IV calcium to stabilize
metabolic status.
Outcome after surgery
• <5% fail to become normocalcaemic, and these should be considered
for a second operation.
• All patients with hyperplasia (including MEN) identified on
histopathology should have long-term monitoring of serum calcium to
detect recurrent primary hyperparathyroidism.
• Patients rendered permanently hypoparathyroid by surgery require
lifelong supplements of active metabolites of vitamin D with calcium.
This can lead to hypercalciuria, and the risk of stone formation may
still be present in these patients. Target serum calcium in these
patients should be towards the low end of the reference range to
minimize hypercalciuria.
OTHER CAUSES OF HYPERCALCAEMIA 469
Familial hypocalciuric
hypercalcaemia (FHH)
FHH—also known as familial benign hypercalcaemia. Three types
exist: FHH 1–3 (see Table 6.5).
• Very rare cause of hypercalcaemia.
• Autosomal dominant, with virtually complete penetrance, but may also
be autosomal recessive.
• Mutation in the calcium-sensing receptor which reduces its sensitivity
such that the body behaves as if it were experiencing normocalcaemia,
even though the serum calcium level is elevated.
• Generally benign and is not usually associated with symptoms or
adverse effects, such as renal stones or bone disease.
• Does not usually show any sustained benefit from parathyroidectomy.
• The homozygous state produces severe life-threatening hypercalcaemia
soon after birth (neonatal severe hyperparathyroidism). In such cases,
total parathyroidectomy is lifesaving.
• See Box 6.8 for management of hypercalcaemia.
Patients have low urine calcium excretion (24h <2.5mmol, fasting calcium/
creatinine excretion ratio <0.01 (Box 6.2, p. 457)).
Further reading
Lietman SA, et al. (2009). A novel loss-of-function mutation, Gln459Arg, of the calcium-sensing
receptor gene associated with apparent autosomal recessive inheritance of familial hypocalciuric
hypercalcemia. J Clin Endocrinol Metab 94, 4372–9.
Nesbit MA, et al. (2013). Mutations in AP2S1 cause familial hypocalciuric hypercalcemia type 3. Nat
Genet 45, 93–7.
Nesbit MA, Hannan FM, Howles SA, et al. (2013). Mutations affecting G-protein subunit α11 in
hypercalcemia and hypocalcemia. N Engl J Med 368(26):2476–86.
Pallais JC, Kifor O, Chen YB, et al. (2004). Acquired hypocalciuric hypercalcemia due to autoanti-
bodies against the calcium-sensing receptor. N Engl J Med 351, 362–9.
472 CHAPTER 6 Calcium and bone metabolism
Vitamin D intoxication
• The diagnosis is established by the presence of greatly elevated
concentrations of 25OHD (>125nmol/L) and 1,25(OH)2D, together
with suppressed PTH. If calcitriol or alfacalcidol is the offending
compound, then 25OHD levels will not be elevated.
• In mild cases, particularly when the active vitamin D metabolites are
involved, the only treatment necessary is to withdraw the offending
treatment and let the calcium settle. If the longer-acting vitamin D
metabolites are involved, then active treatment may be necessary.
• Patients should first be stabilized with a saline infusion (see
Box 6.8).
Following this, the traditional management has been to give high-dose oral
glucocorticoid, such as prednisolone 40mg daily, although IV bisphospho-
nates should also be considered in the acute setting.
Sarcoidosis
• Together with other granulomatous disorders, sarcoidosis causes
hypercalcaemia by extrarenal production of 1,25(OH)2D in
granulomata. This process is not under feedback inhibition but is
substrate-regulated. The hypercalcaemia is, therefore, dependent on
vitamin D supply. Patients may present with hypercalcaemia in summer
or following foreign holidays when the endogenous production of
vitamin D is maximal.
• The biochemical picture is of normal 25OHD, raised 1,25(OH)2D, and
suppressed PTH. In addition, other markers of sarcoid activity, such
as raised angiotensin-converting enzyme (ACE) activity, are frequently
present.
• Treatment with high-dose glucocorticoids is recommended to
control sarcoid activity and to minimize the GI effects of the excess
1,25(OH)2D.
Further reading
Bilezikian JP, Watts JT Jr, Fuleihan Gel-H, et al. (2002). Summary statement from a workshop on
asymptomatic primary hyperparathyroidism: a perspective for the 21st century. J Clin Endocrinol
Metab 87, 5353–61.
Marcocci C, et al. (2011). Primary hyperparathyroidism. N Engl J Med 365, 2389–97.
Palazzo FF, Sadler GP (2004). Minimally invasive parathyroidectomy. BMJ 328, 849–50.
Peacock M, Bilezikian JP, Klassen PS, et al. (2005). Cinacalcet hydrochloride maintains long-term
normocalcemia in patients with primary hyperthyroidism. J Clin Endocrinol Metab 90, 135-41.
Stewart AF (2005). Clinical practice. Hypercalcemia associated with cancer. N Engl J Med
352, 373–9.
Yu N, et al. (2011). A record linkage study of outcomes in patients with mild primary hyperpar-
athyroidism: the Parathyroid Epidemiology and Audit Research Study (PEARS). Clin Endocrinol
(Oxf) 75,169–76.
SARCOIDOSIS 473
474 CHAPTER 6 Calcium and bone metabolism
Hypocalcaemia
Causes
Although hypocalcaemia can result from failure of any of the mechanisms
by which serum calcium concentration is maintained, it is usually the result
of either failure of PTH secretion or because of the inability to release
calcium from bone. These causes are summarized in Box 6.9.
Clinical features
The clinical features of hypocalcaemia are largely as a result of i neuro-
muscular excitability. In order of i severity, these include:
• Tingling—especially of fingers, toes, or lips.
• Numbness—especially of fingers, toes, or lips.
• Cramps.
• Carpopedal spasm.
• Stridor due to laryngospasm.
• Seizures.
The symptoms of hypocalcaemia tend to reflect the severity and rapidity
of onset of the metabolic abnormality.
Clinical signs of hypocalcaemia depend upon the demonstration of neu-
romuscular irritability before this necessarily causes symptoms:
• Chvostek’s sign is elicited by tapping the facial nerve in front of the
ear. A +ve result is indicated by twitching of the corner of the mouth.
Slight twitching is seen in up to 15% of normal ♀, but more major
involvement of the facial muscles is indicative of hypocalcaemia or
hypomagnesaemia.
• Trousseau’s sign is produced by occlusion of the blood supply to the
arm by inflation of a sphygmomanometer cuff above arterial pressure
for 3min. If +ve, there will be carpopedal spasm which may be
accompanied by painful paraesthesiae.
In addition, there may be clinical signs and symptoms associated with the
underlying condition:
• Vitamin D deficiency may be associated with generalized bone pain,
fractures, or proximal myopathy (b see p. 484).
• Hypoparathyroidism can be accompanied by mental slowing and
personality disturbances as well as extrapyramidal signs, cataracts, and
papilloedema.
• If hypocalcaemia is present during the development of permanent
teeth, these may show areas of enamel hypoplasia. This can be a useful
physical sign, indicating that the hypocalcaemia is long-standing.
HYPOCALCAEMIA 475
Pseudohypoparathyroidism
• Resistance to parathyroid hormone action.
• Due to defective signalling of PTH action via cell membrane receptor.
• Also affects TSH, LH, FSH, and GH signalling.
• Most commonly caused by autosomal dominant mutation of
GNAS1 gene.
• Significant imprinting:
• Maternal transmission leads to full blown syndrome of hormone
resistance.
• Paternal transmission causes only phenotypic features of Albright’s
hereditary osteodystrophy.
476 CHAPTER 6 Calcium and bone metabolism
Treatment of hypocalcaemia
• Acute symptomatic hypocalcaemia is a medical emergency and demands
urgent treatment whatever the cause (see Box 6.10).
• Treatment of chronic hypocalcaemia is more dependent on the cause.
Chronic hypocalcaemia
Hypoparathyroidism
• In hypoparathyroidism, the target serum calcium should be at the
low end of the reference range. The reason for this is that the renal
retention of calcium brought about by PTH has been lost. Thus,
any attempt to raise the plasma calcium well into the normal range
is likely to result in unacceptable hypercalciuria, with the risk of
nephrocalcinosis and renal stones.
• In patients with mild parathyroid dysfunction, it may be possible
to achieve acceptable calcium concentrations by using calcium
supplements alone. If used in this way, these need to be given in large
doses, perhaps as much as 1g elemental calcium 3× daily.
• The majority of patients will not achieve adequate control with such
treatment. In those cases, it is necessary to use vitamin D or its
metabolites in pharmacological doses to maintain plasma calcium. The
more potent analogues of vitamin D, such as calcitriol or alfacalcidol,
have the advantage over high-dose calciferol that it is easier to
make changes in therapy in response to plasma calcium levels. If
hypercalcaemia does occur, it settles much more quickly following
withdrawal of these compounds than calciferol. The dose of vitamin D
metabolite is determined by the clinical response but usually lies in the
range of 0.5–2 micrograms daily of one of the potent activated vitamin
D analogues. Serum calcium must be checked on an ongoing basis, and
close monitoring should occur around times of dose adjustments.
• It is essential to ensure an adequate intake of calcium as well as
appropriate doses of activated vitamin D analogues. In some patients,
it is necessary to give calcium supplementation, particularly in
the young.
• PTH (1-84) is a not yet approved therapy but may improve quality of
life.
Pseudohypoparathyroidism
• The principles underlying the treatment of pseudohypoparathyroidism
are the same as those underlying hypoparathyroidism.
• Patients with the most common form of pseudohypoparathyroidism
may have resistance to the action of other hormones which rely on
G protein signalling. They, therefore, need to be assessed for thyroid
and gonadal dysfunction (because of defective TSH or gonadotrophin
action). If these deficiencies are present, they need to be treated in the
conventional manner.
Vitamin D deficiency
Treatment of osteomalacia and vitamin D deficiency is described on
b see p. 484.
TREATMENT OF HYPOCALCAEMIA 479
Vitamin D deficiency
Causes
• Poor sunlight exposure:
• Elderly housebound.
• Extensive skin coverage with clothes.
• Poor diet (especially vegetarians):
• Malabsorption.
• i catabolism of vitamin D.
• s hyperparathyroidism:
• Malabsorption.
• Post-gastrectomy.
• Enzyme-inducing drugs, e.g. phenytoin.
Investigation
The diagnosis of vitamin D deficiency is based on the characteristic bio-
chemical abnormalities (see Table 6.6). Frank osteomalacia is usually asso-
ciated with very low levels of 25OHD (<15nmol/L), but the associated
s hyperparathyroidism frequently results in normal, or even elevated,
concentrations of 1,25(OH)2D.
Treatment
• Treatment is best given in the form of calciferol to restore body
stores, correct biochemical abnormalities, and heal bony abnormalities.
Although the use of the active metabolites of vitamin D will heal the
bony abnormalities, it will not correct the underlying biochemical
problem and is associated with i risk of hypercalcaemia.
• In adults, treatment can be given as a daily dose of calciferol (800–
1,000IU) which is often most easily administered in combination with
a calcium supplement. An alternative, which is particularly helpful if
poor compliance is suspected, is to give a single large dose of 150,000–
300,000IU. This is most effectively given as a single oral dose (3–6 ×
1.25mg tablets of ergocalciferol) which can be supervised in clinic. It is
possible to give a similar dose by IM injection, but the absorption from
this route is variable.
• Following treatment, there is usually a rapid improvement of myopathy
and symptoms of hypocalcaemia. Bone pain frequently persists longer,
and biochemical abnormalities may not settle for several months.
Indeed, following the onset of therapy, markers of bone turnover, such
as alkaline phosphatase, might even show a transient increase, as the
osteoid is mineralized and remodelled.
X-linked hypophosphataemia
• X-linked dominant genetic disorder.
• Severe rickets and osteomalacia.
• Mutation of an endopeptidase gene (PHEX).
Clinical features
• The abnormal phosphate levels are often detected early in infancy, but
skeletal deformities are not apparent until walking commences.
VITAMIN D DEFICIENCY 485
Hypophosphataemia
Phosphate is important for normal mineralization of bone. In the absence
of sufficient phosphate, osteomalacia results. Clinically, osteomalacia is
often indistinguishable from other causes, although there may be features
that will help distinguish the underlying cause of hypophosphataemia. In
addition, phosphate is important in its own right for neuromuscular func-
tion, and profound hypophosphataemia can be accompanied by encepha-
lopathy, muscle weakness, and cardiomyopathy. It must be remembered
that, as phosphate is primarily an intracellular anion, a low plasma phos-
phate does not necessarily represent actual phosphate depletion. Several
different causes of hypophosphataemia are recognized (see Box 6.13).
Treatment
• Mainstay is phosphate replacement, usually Phosphate-Sandoz®, each
tablet of which provides 500mg of phosphate.
• Ideally, patients should receive 2–3g of phosphate daily between
meals, but this is not easy to achieve. All phosphate preparations are
unpalatable and act as osmotic purgatives, causing diarrhoea.
• Long-term administration of phosphate supplements stimulates
parathyroid activity. This can lead to hypercalcaemia, a further
fall in phosphate, with worsening of the bone disease due to the
development of hyperparathyroid bone disease which may necessitate
parathyroidectomy.
• To minimize parathyroid stimulation, it is usual to give one of the
active metabolites of vitamin D in conjunction with phosphate.
Typically, alfacalcidol or calcitriol in a dose of 1–2 micrograms daily
is used.
• Patients receiving such supraphysiological doses of vitamin D
metabolites are at continued risk of hypercalcaemia and require
regular monitoring of plasma calcium, preferably at least every
3 months. The adequacy of calcitriol replacement can be assessed by
maintaining 24h urinary calcium excretion >4–6mmol/day.
• In adults, the role of treatment for X-linked hypophosphataemia is
probably confined to symptomatic bone disease.
• There is little evidence that it will improve the long-term outcome.
• There has even been some evidence that treatment might
accelerate new bone formation.
• In children, treatment is usually given in the hope of improving final
height and minimizing skeletal abnormality—the evidence that it is
possible to achieve these goals is conflicting.
HYPOPHOSPHATAEMIA 487
Further reading
Bergwitz C, Collins MT, Kamath RS et al. (2011) A 56-year old with hypophosphataemea NEJM
365, 1625–35.
Liamis G, Milionis HJ, Elisaf M et al. (2010) Medication-induced hypophosphatemia. QJM 103,
449–59.
488 CHAPTER 6 Calcium and bone metabolism
Hypomagnesaemia
Introduction
• Low plasma magnesium levels are common in acutely ill patients.
Clinical manifestations of this are less common. The most common
clinical feature of magnesium deficiency is neuromuscular excitability,
muscular weakness which is virtually indistinguishable from that
associated with hypocalcaemia, which frequently coexists. Arrhythmias
can also occur, as can coma.
• Positive Chvostek’s and Trousseau’s signs are seen.
• Is associated with hypocalcaemia and hypokalaemia.
• In the presence of magnesium deficiency, PTH secretion is inhibited
and the peripheral action of PTH is also attenuated. Resultant
hypomagnesaemia-induced hypocalcaemia will not respond to
calcium treatment, unless the magnesium deficiency is corrected
first. Hypokalaemia is also frequently seen in association with
hypomagnesaemia. Treatment with potassium supplementation is often
unsuccessful, unless magnesium is replaced at the same time.
• See Box 6.14 for causes.
Treatment
Symptomatic magnesium deficiency, especially if associated with hypocal-
caemia or hypokalaemia, requires parenteral treatment.
• Magnesium sulfate 50% solution contains 72mmol magnesium/mL. This
should be administered IV, although IM is feasible but painful. An initial
4–8mmol should be given over 15min, followed by a slow infusion
of 1mmol/h. The rate of the infusion can be adjusted in light of the
response in plasma magnesium.
• In the presence of renal impairment, plasma magnesium can rise
quickly, and so particular care must be undertaken if magnesium
infusion is contemplated in the presence of renal failure.
• After repletion has been achieved intravenously, or in less severe
cases, treatment can be continued orally.
• Various salts of magnesium have been used, including chloride, oxide,
and glycerophosphate. Dosing is frequently limited by the purgative
properties of magnesium salts.
HYPOMAGNESAEMIA 489
Further reading
Ayuk J, Gittoes NJ (2011). How should hypomagnesaemia be investigated and treated? Clin
Endocrinol (Oxf) 75, 743–6.
490 CHAPTER 6 Calcium and bone metabolism
Osteoporosis
Introduction
Although the term osteoporosis refers to the reduction in the amount
of bony tissue within the skeleton, this is generally associated with a loss
of structural integrity of the internal architecture of the bone. The com-
bination of both these changes means that osteoporotic bone is at high
risk of fracture, even after trivial injury. The most common osteoporotic
fractures are those of the hip, wrist (Colles’), and compression fractures of
vertebral bodies. Patients who fracture ribs, the upper humerus, leg, and
pelvis also have a higher incidence of osteoporosis.
Over recent years, there has been a change in focus in the treatment
of osteoporosis. Historically, there has been a primary reliance on bone
mineral density as a threshold for treatment, whereas currently there is
far greater emphasis on assessing individual patients’ risk of fracture that
incorporates multiple clinical risk factors as well as bone mineral density.
See Box 6.15 for causes.
Pathology of osteoporosis
Osteoporosis may arise from a failure of the body to lay down sufficient
bone during growth and maturation; an earlier than usual onset of bone
loss following maturity; or an i rate of that loss.
See Table 6.7 for definitions and Box 6.16 for causes.
Peak bone mass
• Mainly genetically determined:
• Racial effects (bone mass higher in Afro-Caribbean and lower in
Caucasians).
• Family influence on the risk of osteoporosis—may account for 70%
of variation.
• So far, >9 separate genetic associations described.
• Also influenced by environmental factors:
• Exercise—particularly weight-bearing.
• Nutrition—especially calcium.
• Exposure to oestrogen is also important:
• Early menopause or late puberty (in ♂ or ♀) is associated with
i risk of osteoporosis.
Early onset of loss
• Early menopause.
• Conditions leading to bone loss, e.g. glucocorticoid therapy.
Increased net loss
• Ageing:
• Vitamin D insufficiency.
• Declining bone formation.
• Declining renal function.
• Underlying disease states (see Box 6.15).
Lifestyle factors affecting bone mass
Increase
• Weight-bearing exercise.
Decrease
• Smoking.
• Excessive alcohol.
• Nulliparity.
• Poor calcium nutrition.
PATHOLOGY OF OSTEOPOROSIS 493
Table 6.7 WHO definitions for osteoporosis and low bone mass
T score Fragility fracture Diagnosis
–1 Normal
<–1 but ≥–2.5 Low bone mass (osteopenia)
<–2.5 No Osteoporosis
<–2.5 Yes Established (severe) osteoporosis
Epidemiology of osteoporosis
• The risk of osteoporotic fracture increases with age. Fracture rates in
♂ are approximately half of those seen in ♀ of the same age. A ♀
aged 50 has approximately a 1:2 chance of sustaining an osteoporotic
fracture in the rest of her life. The corresponding figure for a ♂ is 1:5.
• In the UK each year, in ♀, there are in excess of 25,000 vertebral
fractures that come to clinical attention, together with over 40,000
wrist fractures and 50,000 hip fractures. The latter are a particular
health challenge, as they invariably result in hospital admission.
One-fifth of hip fracture victims will die within 6 months of the injury,
and only 50% will return to their previous level of independence. It has
been estimated that the overall cost of osteoporotic fractures in the
UK is £2.1 billion annually.
• See Box 6.17 for investigations.
Presentation of osteoporosis
• Usually clinically silent until an acute fracture.
• Two-thirds of vertebral fractures do not come to clinical attention.
• Typical vertebral fracture:
• Sudden episode of well-localized pain.
• May, or may not, have been related to injury or exertion.
• May be radiation of the pain in a girdle distribution.
• Pain may initially require bed rest but gradually subsides over
4–8 weeks; even after this time, there may be residual pain at the
fracture site.
• Osteoporotic vertebral fractures only rarely lead to neurological
impairment.
• Any evidence of spinal cord compression should prompt a search for
malignancy or other underlying cause.
• Following vertebral fracture, a patient may be left with persistent back
pain, kyphosis, or height loss.
• Although height loss and kyphosis are often thought of as being
indicative of osteoporosis, they are more frequently the result of
degenerative disease, including disc disease. These changes cannot be
attributed to osteoporosis in the absence of vertebral fractures.
• Peripheral fractures are also more common in osteoporosis.
• If a bone breaks from a fall from less than standing height, that
represents a low-trauma fracture which might indicate underlying
osteoporosis.
• Osteoporosis does not cause generalized skeletal pain.
496 CHAPTER 6 Calcium and bone metabolism
Investigation of osteoporosis
Establish the diagnosis
• Plain radiographs are useful for determining the presence of
fracture. Apart from this, they are of little utility in the diagnosis of
osteoporosis. Bone density cannot be assessed reliably from a plain
radiograph.
• Bone densitometry. In order to identify the presence of T score-defined
osteoporosis, it is important to measure BMD appropriately. This
is usually carried out at the hip and lumbar spine, using dual-energy
X-ray absorptiometry (DXA). The presence of degenerative disease
or arterial calcification can elevate the apparent bone density of the
spine without adding to skeletal strength. i reliance is, therefore, being
placed on measurements derived from the hip. The diagnostic criteria
for osteoporosis have been derived through the WHO (see Table 6.7).
• Biochemical markers of bone turnover may be helpful in the
calculation of fracture risk and in judging the response to drug
therapies, but they have no role in the diagnosis of osteoporosis.
• Use of fracture risk algorithms is now available to determine
individuals’ fracture risk (e.g. M http://www.shef.ac.uk/FRAX). Such an
approach can help tune therapies to those patients at heightened risk
of fracture.
Exclude underlying causes
An underlying cause for osteoporosis is present in approximately 10–30%
of women and up to 50% of men with osteoporosis. Many of the underly-
ing causes (see Box 6.15) should be apparent from a careful history and
physical examination. A few basic investigations are useful to exclude the
more common underlying causes. Other investigations may be needed to
exclude other specific conditions.
It is helpful to target investigations at those people who have a signifi-
cantly lower than expected bone mass for their age (Z score <–2).
Monitoring therapy
• Repeat bone densitometry is not obligatory for monitoring, as there
is no direct correlation between changes in bone density and the
anti-fracture effects of drugs. This is an area of controversy.
• Minimum time interval should be 2 years between scans.
• Effective treatment leads to modest rise (75% at spine) in bone density.
• Biochemical markers of bone turnover may be helpful in specific
settings (e.g. P1NP and NTX).
• See Table 6.8 for efficacy for types of treatments
INVESTIGATION OF OSTEOPOROSIS 497
Treatment of osteoporosis
Treatments should be considered according to their effect on fracture risk
reduction and side effect profile.
In addition to pharmacological treatments aimed at reducing fracture
risk, it must be remembered that non-pharmacological measures are also
important. Thus, it is prudent to minimize the risk of falling by adjusting
the home environment and reviewing the need for medications, such as
hypnotics and antihypertensives, for instance.
Lifestyle measures
• Stop smoking.
• Avoid alcohol excess.
• Encourage weight-bearing exercise:
• Lower-impact exercise, e.g. walking outdoors for 20min 3× weekly,
may reduce fracture risk.
• Encourage well-balanced diet.
• Ensure adequate calcium and vitamin D intake.
• If necessary, give supplements to achieve calcium intake of 71g daily.
Choice of therapy and who to treat
The National Institute for Health and Care Excellence (NICE) has
produced guidance on the s prevention of osteoporotic fractures in
post-menopausal women with a low-trauma fracture (M http://guidance.
nice.org.uk/TA161; M http://guidance.nice.org.uk/TA204). The guidelines
focus on age, bone density, clinical risk factors, and cost effectiveness to
derive recommendations.
The National Osteoporosis Guideline Group (NOGG) has also derived
alternative clinical guidelines that are based on a fracture risk assess-
ment tool (FRAX) that links into a web-based guideline tool to derive
recommendations regarding prescribing (M http://www.shef.ac.uk/FRAX;
M http://www.shef.ac.uk/NOGG).
Therapy is given for 5 years in the first instance. Reassessment should
occur at this time. If the T score at the femoral neck is below –2.5, then
therapy should be continued. Consideration should also be given to con-
tinue therapy if there has been a previous vertebral fracture. Patients with
a T score above –2.0 at the femoral neck are unlikely to benefit from
continued treatment.
See Box 6.18 for vertebroplasty.
Glucocorticoid-induced osteoporosis
Glucocorticoid treatment is one of the major s causes of osteoporosis.
Not all patients receiving steroid treatment do lose bone, and it is not
clear what determines this. Patients who sustain fractures while taking glu-
cocorticoids do so at higher bone density levels than in post-menopausal
women, leading to the assertion that such patients should be treated at a
higher bone density (T <–1.5) than would be the case for post-menopausal
osteoporosis.
The Royal College of Physicians (UK) has produced guidelines for the
management of glucocorticoid-induced osteoporosis (see Fig. 6.4).
TREATMENT OF OSTEOPOROSIS 499
Strontium ranelate
• Diarrhoea.
• Venous thromboembolism.
Denosumab
• Effective for at least 6 years.
• Skin infections/eczema.
• Hypocalcaemia (ensure Ca and vitamin D levels normal prior).
Teriparatide
• Contraindications: hypercalcaemia, renal impairment, unexplained
elevation of alkaline phosphatase—Paget’s disease, prior irradiation.
• Risk of hypercalcaemia is not great. and no specific monitoring of
treatment is recommended.
• Vomiting.
• Leg cramps.
• i risk of osteosarcoma seen in rats given teriparatide for most of
their life. It should be avoided in patients with i risk of bone tumours
(Paget’s disease, raised alkaline phosphatase, previous skeletal
radiotherapy).
Causes of increased bone mineral density
Artefacts
• Excess skeletal calcium (lumbar spondylosis, ankylosing spondylitis).
• Extraskeletal calcium (vascular, gallstones).
• Vertebral fracture.
• Radiodense material (surgical implants, strontium).
Focal increase in bone
• Paget’s disease.
• Tumours (e.g. haemangioma, Hodgkin’s, plasmacytoma).
Generalized increase in bone
Acquired osteosclerosis (general osteodystrophy, fluorosis, mastocytosis,
hepatitis C, myelofibrosis, acromegaly).
Genetic sclerosing bone dysplasias
• d absorption (e.g. osteopetrosis).
• i formation (e.g. sclerosteosis).
• Disturbed balance of formation and resorption.
504 CHAPTER 6 Calcium and bone metabolism
Further reading
Black DM, Delmas PD, Eastell R, et al. (2007). Once-yearly zoledronic acid for treatment of post-
menopausal osteoporosis. N Engl J Med 356, 1809–22.
Boonen S, et al. (2012). Fracture risk and zoledronic acid therapy in men with osteoporosis. N Engl
J Med 367, 1714–23.
Ebeling PR (2008). Osteoporosis in men. N Engl J Med 358, 1474–82.
Khosla S, et al. (2008). Osteoporosis in men. Endocr Rev 29, 441–64.
Klazen CA, et al. (2010). Vertebroplasty versus conservative treatment in acute osteoporotic ver-
tebral compression fractures (Vertos II): an open-label randomised trial. Lancet 376, 108–92.
Meunier PJ, Roux C, Seeman E, et al. (2004). The effects of strontium ranelate on the risk of
vertebral fracture in women with postmenopausal osteoporosis. N Engl J Med 350, 459–68.
Ranney A, Tugwell P, Wells G, et al. (2002). Meta-analyses of therapies for postmenopausal osteo-
porosis. I. Systematic reviews of randomized trials in osteoporosis: induction and methodology.
Endocr Rev 23, 496–507.
Russell RGG (2011). Bisphosphonates: the first 40 years. Bone 49, 2–19.
Sambrook P, Cooper C (2006). Osteoporosis. Lancet 367, 2010–28.
Schilcher J, et al. (2011). Bisphosphonate use and atypical fractures of the femoral shaft. N Engl J
Med. 364, 1728–37.
Watts NB, at al. (2012). Osteoporosis in men: an Endocrine Society clinical practice guideline. J Clin
Endocrinol Metab 97, 1802–22.
Weinstein RS (2011). Clinical practice. Glucocorticoid-induced bone disease. N Engl J Med
365, 62–70.
506 CHAPTER 6 Calcium and bone metabolism
Paget’s disease
Paget’s disease is the result of greatly i local bone turnover, which occurs
particularly in the elderly but can affect younger people.
Pathology
• The p abnormality in Paget’s disease is gross overactivity of the
osteoclasts, resulting in greatly i bone resorption. This secondarily
results in i osteoblastic activity. The new bone is laid down in a highly
disorganized manner and leads to the characteristic pagetic abnormality,
with irregular packets of woven bone being apparent on biopsy and
disorganized internal architecture of the bone on plain radiographs.
• Paget’s disease can affect any bone in the skeleton but is most
frequently found in the pelvis, vertebral column, femur, skull, and tibia.
In most patients, it affects several sites, but, in about 20% of cases, a
single bone is affected (monostotic disease). Typically, the disease will
start in one end of a long bone and spread along the bone at a rate of
about 1cm per year. Although it can spread within an affected bone,
it appears that the pattern of disease is fixed by the time of clinical
presentation, and it is exceedingly rare for new bones to become
involved during the course of the disease.
• Paget’s disease alters the mechanical properties of the bone. Thus,
pagetic bones are more likely to bend under normal physiological
loads and are thus liable to fracture. This can take the form of
complete fractures, which tend to be transverse, rather than the more
common spiral fractures of long bones. More frequently, fissure or
incremental fractures are seen on the convex surface of bowed pagetic
bones. These may be painful in their own right but are also liable to
proceed to complete fracture. Pagetic bones are also larger than their
normal counterparts. This can lead to i arthritis at adjacent joints and
to pressure on nerves, leading to neurological compression syndromes
and, when it occurs in the skull base, sensorineural deafness.
Aetiology of Paget’s disease
Unclear. There are two major theories:
• Familial:
• Some genetic associations, especially with a sequestasome-1
mutation.
• These are not invariable.
• Viral:
• Inclusion bodies, similar to those seen in viral infections, have been
identified in osteoclasts from patients with Paget’s.
• Some workers have found paramyxoviral (measles or canine
distemper) protein or nucleic acid in pagetic bone; others have not
been able to replicate this.
PAGET’S DISEASE 507
Epidemiology
Paget’s disease is present in about 2% of the UK population over the
age of 55. Its prevalence increases with age, and it is more common in
♂ than ♀. Only about 10% of affected patients will have symptomatic
disease. It is most common in the UK or in migrants of British descent
in North America and Australasia but rare in Africa. Studies in the UK
and New Zealand have suggested that the prevalence may be declining
with time.
Clinical features
• 90% asymptomatic.
• Most notable feature is pain. This is frequently multifactorial:
• i metabolic activity of the bone.
• Changes in bone shape.
• Fissure fractures.
• Nerve compression.
• Arthritis.
• Pagetic bones tend to increase in size or become bowed (16%
cases): bowing can be so severe as to interfere with function.
• Fractures (either complete or fissure) present in 10%.
• Risk of osteosarcoma is increased in active Paget’s disease but is a very
rare finding.
Investigation
The diagnosis of Paget’s disease is primarily radiological.
Radiological features of Paget’s disease
• Early disease—primarily lytic:
• V-shaped ‘cutting cone’ in long bones.
• Osteoporosis circumscripta in skull.
• Combined phase (mixed lytic and sclerotic):
• Cortical thickening.
• Loss of corticomedullary distinction.
• Accentuated trabecular markings.
• Late phase—primarily sclerotic:
• Thickening of long bones.
• Increase in bone size.
• Sclerosis.
An isotope bone scan is frequently helpful in assessing the extent of skel-
etal involvement with Paget’s disease. It is particularly important to identify
Paget’s disease in a weight-bearing bone because of the risk of fracture.
The uptake of tracer depends on the disease activity, and isotope bone
scans can also be used to assess the response to therapy.
In active disease, plasma alkaline phosphatase activity is usually (85%)
elevated. An exception to this is in monostotic disease when there may
be insufficient bone involved to raise the enzyme levels above normal.
Alkaline phosphatase activity responds to successful treatment. There is
little advantage in using the more modern markers of bone turnover over
the total alkaline phosphatase activity for the monitoring of pagetic activity.
A possible exception to this is in patients with liver disease where changes
in bone alkaline phosphatase might be masked by the liver isoenzyme.
508 CHAPTER 6 Calcium and bone metabolism
Complications
• Deafness is present in up to half of cases of skull base Paget’s.
• Other neurological complications are rare. These can include:
• Compression of other cranial nerves with skull base disease.
• Spinal cord compression. Most common with involvement of the
thoracic spine and is thought to result as much from a vascular steal
syndrome as from physical compression. It frequently responds to
medical therapy without need for surgical decompression.
• Platybasia which can lead to an obstructive hydrocephalus that may
require surgical drainage.
• Osteogenic sarcoma:
• Very rare complication of Paget’s disease.
• Rarely amenable to treatment.
• Presents with i pain/radiological evidence of tumour, a mass, and
very elevated alkaline phosphatase.
• Any increase of pain in a patient with Paget’s disease should arouse
suspicion of sarcomatous degeneration. A more common cause,
however, is resumption of activity of disease.
Pagetic sarcomas are most frequently found in the humerus or femur but
can affect any bone involved with Paget’s disease.
Treatment
Treatment with agents that decrease bone turnover reduces disease
activity, as indicated by bone turnover markers and isotope bone scans.
There is evidence to suggest that such treatment leads to the deposi-
tion of histologically normal bone. Although such treatment has been
shown to help pain, there is little evidence that it benefits the other con-
sequences of Paget’s disease. In particular, the deafness of Paget’s disease
does not regress after treatment, although its progression may be halted.
Nonetheless, it has become generally accepted to treat patients in the
hope that future complications of the disease will be avoided. Typical indi-
cations for treatment of Paget’s disease are listed in Box 6.21.
Bisphosphonates have become the mainstay of treatment. IV zoledro-
nate is the drug of choice and results in long-term normalization of alkaline
phosphatase in the majority of patients. Calcitonin and plicamycin are no
longer used.
Goals of treatment
• Minimize symptoms.
• Prevent long-term complications.
• Normalize bone turnover.
• Alkaline phosphatase in normal range.
• No actual evidence that treatment achieves this.
PAGET’S DISEASE 509
Monitoring therapy
• Plasma alkaline phosphatase every 6–12 months.
• Clinical assessment.
Re-treat if symptoms recur with objective evidence of disease recurrence
(alkaline phosphatase or +ve isotope scan). There is no evidence that
treating a raised alkaline phosphatase in the absence of symptoms affects
outcome in Paget’s.
Further reading
Albagha OM, et al. (2011). Genome-wide association identifies three new susceptibility loci for
Paget’s disease of bone. Nat Genet 43, 685–9.
Ralston SH (2013). Clinical practice. Paget’s disease of bone. N Engl J Med 368, 644–50.
Ralston SH, et al (2008). Pathogenesis and management of Paget’s disease of bone. Lancet 372,
155–163.
Selby PL, Davie MW, Ralston SH, et al. (2002). National Association for the Relief of Paget’s
Disease: guidelines on the management of Paget’s disease of the bone. Bone 31, 366–73.
510 CHAPTER 6 Calcium and bone metabolism
Further reading
Bitton A, et al. (2009). Clinical problem-solving. A fragile balance. N Engl J Med 361, 74–9.
Rauch F, Glorieux FH (2004). Osteogenesis imperfecta. Lancet 363, 1377–85.
Chapter 7 513
Paediatric endocrinology
Growth 514
Short stature 518
Constitutional delay of growth and puberty 520
Primary GH deficiency 522
Secondary GH deficiency 524
Treatment of GH deficiency 526
GH resistance 528
Hypothyroidism 529
Coeliac disease 530
Skeletal dysplasias 530
Small for gestational age (SGA) and intrauterine growth
restriction 531
Turner’s syndrome 532
Tall stature and rapid growth 534
Normal puberty 536
Precocious puberty 538
Delayed/absent puberty 542
Normal sexual differentiation 544
Assessment of ambiguous genitalia 545
Disorders of sex development (DSD) 546
Congenital adrenal hyperplasia 550
514 CHAPTER 7 Paediatric endocrinology
Growth
Regulation of growth
Normal human growth can be divided into three overlapping stages (the
Karlberg model), each under the control of different factors:
• Infancy. Growth is largely under nutritional regulation, and wide
inter-individual variation in rates of growth is seen. Many infants show
significant ‘catch-up’ or ‘catch-down’ in weight and length, and by
2 years, length is much more predictive of final adult height than at birth.
• Childhood. Growth is regulated by growth hormone (GH) and thyroxine.
It is characterized by alternating periods of mini-growth spurts with
intervening stasis, each phase lasting several weeks. However, over
years, a child will tend to maintain their centile position on height charts,
with a height velocity between the 25th and 75th centiles.
• Puberty. The combination of GH and sex hormones promotes bone
maturation and a rapid growth acceleration or ‘growth spurt’. In both
sexes, oestrogen eventually causes epiphyseal fusion, resulting in the
attainment of final height.
Sex differences
Adult heights differ between ♂ and♀ by, on average, 13cm. However,
during childhood, onset of the pubertal growth spurt is earlier in ♀, who
are therefore, on average, taller than ♂ between the ages of 10–13 years.
Tempo
Within each sex, there may also be marked inter-individual differences
in tempo of growth (or rate of attainment of final height) and the timing
of puberty. Delay or advance of bone maturation is linked with timing of
puberty. Constitutional delay in growth and puberty often runs in families,
reflecting probable genetic factors. Comparison of bone age (estimated
from a hand radiograph) with chronological age is, therefore, an important
part of growth assessment.
Final height
Final height is estimated as the height reached when growth velocity
slows to <2cm/year and can be confirmed by finding epiphyseal fusion
on hand radiograph ± knee radiograph. Final height is largely genetically
determined, and a target height can be estimated in each individual from
their parent’s heights.
Assessment of growth
Measurement
• From birth to 2 years old, supine length is measured ideally using a
measuring board (e.g. Harpenden neonatometer). Two adults are
needed to ensure that the child is lying straight and legs extended.
• From 2 years old, standing height is measured against a wall-mounted
or free-standing stadiometer, with the measurer applying moderate
upwards neck traction and the child looking forward in the
horizontal plane.
GROWTH 515
Secular trends
Children’s heights i by >1cm in England and by >2cm in Scotland during
the period from 1972–94, and similar trends are seen in many other coun-
tries. Population growth references used should be appropriate to the
population studied and may occasionally need to be updated. However,
secular trends in height over the last decade have reduced significantly in
some countries.
190
180 Boys
170
Girls
160
150
140
130
Height (cm)
120
110
100
90
80
70
60
50
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19
Age (years)
Fig. 7.1 Typical individual height-attained curves for boys and girls (supine length
to the age of 2; integrated curves of Fig. 7.2).
GROWTH 517
24
23
22
21
20
19
18
17
16
15
14
Height (cm)
13
12
11
10
Boys
9
Girls
8
7
6
5
4
3
2
1
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19
Age (years)
Fig. 7.2 Typical individual velocity curves for supine length or height in boys and
girls. These curves represent the velocity of the typical boy and girl at any given
instant.
Reference
1. De Waal WJ, Greyn-Fokker MH, Stijnen T, et al. (1996). Accuracy of final height prediction and
effect of growth-reductive therapy in 362 constitutionally tall children. J Clin Endocrinol Metab
81, 1206–16.
518 CHAPTER 7 Paediatric endocrinology
Short stature
Definition
Short stature is defined as height <2nd centile for age and sex on the UK
1990 growth chart. However, abnormalities of growth may be present long
before attained height falls below this level and may be detected much
earlier by assessing growth velocity and observing height measurements.
Assessment
History
• Who is concerned, child or parents?
• What are the parental heights?
• Has the child always been small or does the history suggest recent
growth failure? Try to obtain previous measurements (e.g. from
parents, GP, health visitor, school).
• Ask about maternal illness in pregnancy, drug intake and possible
substance abuse in pregnancy, gestation at delivery, size at birth
(weight/length/head circumference), childhood illnesses, medication,
and developmental milestones.
• Systematic enquiry for headaches, visual disturbance, asthma/
respiratory symptoms, abdominal symptoms, and diet.
• Is there a family history of short stature or pubertal delay?
• What are the psychosocial circumstances of the child and the family?
Examination
• Assess height and height velocity over at least 6 months.
• Measure sitting height, and derive subischial leg length (standing
height – sitting height (cm)) if skeletal disproportion suspected.
• Assess for the presence and severity of chronic disease. Low weight
for height suggests a nutritional diagnosis, GI cause, or other significant
systemic disease.
• Pubertal stage using Tanner’s criteria (see b p. 537).
• Observe for dysmorphic features and signs of endocrinolopathy,
presence and severity of chronic disease.
• Measure parents’ heights, and calculate MPH.
Investigations
• Laboratory tests should include FBC, ESR, electrolytes, thyroid
function, calcium, phosphate, antigliadin and antiendomysial
antibodies, IGF-I level, karyotype (of particular importance in girls),
and urinalysis.
• These tests may also be clinically indicated: GH provocation testing
(e.g. arginine, glucagon, or ITT) (see Table 7.1) with other anterior
pituitary function tests, MRI scan with specific reference to the
hypothalamus and pituitary, skeletal survey (for bone dysplasia),
and, very rarely, an IGF-1 generation test (for GH resistance, b see
Growth hormone deficiency, p. 521).
Causes
• Genetic short stature.
SHORT STATURE 519
Primary GH deficiency
p GH deficiency is usually sporadic, but rarely it may be inherited as
autosomal dominant, recessive, or X-linked recessive and may be associ-
ated with other pituitary hormone deficiencies. It may represent a defect
in homeobox genes (e.g. Pit1 (leading to GH, TSH, and PRL deficiency),
Prop1 (leading to GH, TSH, PRL, gonadotrophin, and later ACTH deficien-
cies), or Hesx1) which control HP development. Mutation in Hesx1 has
been associated with midline defects, such as optic nerve hypoplasia and
corpus callosum defects (i.e. ‘septo-optic dysplasia’). GH deficiency usu-
ally arises because of failure of release of GHRH from the hypothalamus.
Clinical features
• Infancy. GH deficiency may present with hypoglycaemia. Coexisting
ACTH, TSH, and gonadotrophin deficiencies may cause prolonged
hyperbilirubinaemia and micropenis. Size may be normal, as fetal and
infancy growth is more dependent on nutrition and other growth
factors than on GH.
• Childhood. Typical features include slow growth velocity, short stature,
d muscle mass, and i SC fat. Underdevelopment of the mid-facial
bones, relative protrusion of the frontal bones because of mid-facial
hypoplasia, delayed dental eruption, and delayed closure of the
anterior fontanelle may be seen. These children have delayed bone age
and delayed puberty.
PRIMARY GH DEFICIENCY 523
524 CHAPTER 7 Paediatric endocrinology
Secondary GH deficiency
Brain tumours and cranial irradiation
Pituitary or hypothalamic tumours may impair GH secretion, and deficien-
cies of other pituitary hormones may coexist. Cranial irradiation, used to
treat intracranial tumours, facial tumours, and acute leukaemia, may also
cause GH deficiency. Risk of HP damage is related to total dose adminis-
tered, fractionation (single dose more toxic than divided), location of the
irradiated tissue, and age (younger children are more sensitive to radiation
damage).
GH secretion is most sensitive to radiation damage, followed by gon-
adotrophins, TSH, and ACTH. Central precocious puberty may also occur
and may mask GH deficiency by promoting growth but will compromise
final height if untreated. At-risk children should, therefore, be screened
regularly by careful examination and multiple pituitary hormone testing.
These survivors of childhood cancer may also have other endocrine
problems, including gonadal damage related to concomitant chemother-
apy or radiation scatter, hypothyroidism related to spinal radiation, or
glucose intolerance related to total body irradiation. It is recommended
that all such patients should undergo endocrine surveillance.
Psychosocial deprivation
Severe psychosocial deprivation may cause reversible disturbance of GH
secretion and growth failure. GH secretion improves within 3 weeks of
hospitalization or removal from the adverse environment, and catch-up
growth is often dramatic (see Fig. 7.3), although these children may con-
tinue to exhibit other features of emotional disturbance.
SECONDARY GH DEFICIENCY 525
190 97
90
180 BOYS Height 75
50 F
170 97 25
10
Longitudinal 50 3 M
160 standards
3
150
140
130
Height (cm)
120
110
100
90
80
60
50
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19
Age (years)
Fig. 7.3 Height of child with psychosocial short stature. Note catch-up on
removal, marked by arrow, from parental home. Reproduced from Brook C (2001).
Brook’s Clinical Pediatric Endocrinology, Blackwell Publishing. With permission
from Wiley Blackwell.
526 CHAPTER 7 Paediatric endocrinology
Treatment of GH deficiency
(See Fig. 7.4.)
Recombinant human GH has been available since 1985 and is adminis-
tered by daily SC injection.
Dose
The replacement dose for childhood GH deficiency is 25–50 micrograms/
kg/day (70.7–1.4 micrograms/m2 per day). Catch-up growth is optimized if
GH is commenced early. A higher dose can be used in puberty, reflecting
the normal elevation in GH levels at Tanner stage 3–4.
Side effects
Local lipoatrophy and benign intracranial hypertension occur rarely.
Slipped upper femoral epiphyses are associated with GH deficiency, but
the incidence is similar before or after GH treatment. Other pituitary hor-
mone deficiencies may be unmasked by GH therapy, and thyroid function
should be checked within 4–6 weeks of commencing therapy.
Retesting in adulthood
Once final height is achieved, GH secretion should be retested, as a
significant percentage of subjects (25–80%) with GH deficiency in child-
hood subsequently have normal GH secretion in adulthood. In those with
confirmed GH deficiency, continuation of GH treatment (at a dose of
0.2–0.5mg/day) through the late adolescent years into early adulthood
(the transition phase) is recommended in order to complete somatic
development (increasing lean body mass and muscular strength, reducing
fat mass, improving bone density, and maintaining a healthy lipid profile).
GH treatment may need to be continued beyond this phase as adult GH
replacement.
The transition from paediatric to adult care is an important time, not
only to re-evaluate GH status but also to reassess other pituitary function
and management of any underlying disorder.
It is also recommended that assessment of bone mineral density, body
composition, fasting lipid profile, and QoL by questionnaire should be
undertaken at this time and repeated at 3–5-yearly intervals for those
restarting GH treatment.
TREATMENT OF GH DEFICIENCY 527
190 F
97
90
180 BOYS Height 75
50 M
170 97 25
Longitudinal 10
50 3
160 standards
3
150
140
130
Height (cm)
120
110
100
90
80 Penis
stage
70
Pubic hair
60 stage
Testes
50 vol.
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19
Age (years)
Fig. 7.4 Height of one brother with isolated growth hormone deficiency treated
with human GH from age 6 years. Catch-up is partly by high velocity and partly
by prolonged growth and is incomplete. F and M, parents’ height centiles; vertical
thick line, range of expected heights for family. Reproduced from Brook C (2001).
Brook’s Clinical Pediatric Endocrinology, Blackwell Publishing. With permission
from Wiley Blackwell.
528 CHAPTER 7 Paediatric endocrinology
GH resistance
GH resistance may arise because of p GH receptor defects or
post-receptor defects s to malnutrition, liver disease, type 1 diabetes,
or, very rarely, circulating GH antibodies. Laron syndrome is a rare auto-
somal recessive condition caused by a genetic defect of the GH receptor.
Affected individuals have extreme short stature, high levels of GH, low
levels of IGF-I, and impaired GH-induced IGF-I generation (b see Box 7.1,
p. 521). Treatment with recombinant IGF-I is available.
HYPOTHYROIDISM 529
Hypothyroidism
(See Fig. 7.5.)
• Congenital p hypothyroidism is detected by neonatal screening.
• Hypothyroidism presenting in childhood is usually autoimmune in
origin.
• Incidence is higher in girls and those with personal or family history of
other autoimmune disease.
• In childhood, hypothyroidism may present with growth failure alone,
and bone age is often disproportionately delayed. Very rarely, early
puberty may occur.
• Investigations show low T4 and T3, high TSH, +ve antithyroglobulin and
antithyroid peroxidase (microsomal) antibodies.
• Replacement therapy with oral levothyroxine (100 micrograms/m2 per
day, titrated with thyroid function) results in catch-up growth, unless
diagnosis is late.
190
140
130
Height (cm)
120
110
100
Thyroxine
90
80 Breast
stage
70
Pubic hair
60 stage
Menarche
50
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19
Age (years)
Fig. 7.5 Response of hypothyroid child treated with thyroxine. Solid circles,
height for age; open circles, height for bone age. Reproduced from Brook C (2001).
Brook’s Clinical Pediatric Endocrinology, Blackwell Publishing. With permission
from Wiley Blackwell.
530 CHAPTER 7 Paediatric endocrinology
Coeliac disease
More common in children with other autoimmune disorders. Although the
classical childhood presentation is an irritable toddler with poor weight
gain, diarrhoea, abdominal pain, and distension, in later childhood, poor
growth with bone age delay may be the presenting feature. Measurement
of tissue transglutaminase and antiendomysial and antigliadin antibod-
ies are valuable screening tests, but diagnosis needs to be confirmed by
small bowel biopsy. Catch-up growth usually follows commencement of
a gluten-free diet.
Skeletal dysplasias
• This heterogeneous group of mostly dominantly inherited disorders
includes achondroplasia and hypochondroplasia.
• These children usually have severe disproportionate short stature and
a +ve or suspicious family history.
• Radiological assessment by skeletal survey often allows a specific
diagnosis to be made.
• High-dose GH therapy has been used in these disorders, with variable
success. Surgical leg lengthening procedures before and/or after
puberty are also an additional option.
SGA AND INTRAUTERINE GROWTH RESTRICTION 531
Turner’s syndrome
(b also see Box 4.7, p. 337.)
Turner’s syndrome should always be considered in a girl who is short
for her parental target, and the classical dysmorphic features may be dif-
ficult to identify at younger ages. Karyotype usually confirms the diag-
nosis, although sufficient cells (>30) should be examined to exclude the
possibility of mosaicism.
Clinical features
There may be a history of lymphoedema in the newborn period. Typically,
growth velocity starts to decline from 3–5 years old (see Fig. 7.6), and
gonadal failure, combined with a degree of skeletal dysplasia, results in
loss of the pubertal growth spurt. Mean final height is consistently 20cm
below the normal average within each population (143–146cm in the UK).
GH secretion is normal, although IGF-I levels may be low. 10% progress
through puberty spontaneously, but only 1% develop ovulatory cycles.
Management
• High-dose GH therapy (45 micrograms/kg/day) increases final height,
although individual responses are variable. The height gained is related
to time on GH treatment, and thus GH therapy should be commenced
early; if the diagnosis has been made in early life, treatment is usually
started from 3–5 years of age.
• The anabolic steroid oxandrolone can be used, in addition to GH, to
promote growth from the age of 9 years. It has a positive effect on
final height at a dose of 0.05mg/kg/day, max 2.5mg/day.
• Oral oestrogen is commenced between 12–14 years to promote s
sexual development and pubertal growth. It should be started in low
dose (ethinylestradiol 2 micrograms/day) and gradually i with age.
Progesterone should be added if breakthrough bleeding occurs or when
oestrogen dose reaches 10 micrograms/day.
TURNER’S SYNDROME 533
170
160 +2SD
150 +1SD
x
140 −1SD
−2SD
Height (cm)
130
120
110
90
80
70
2 4 6 8 10 12 14 16 18 20
Age (years)
Fig. 7.6 Height SDS for chronological age extrapolated to final height.
Reproduced from Brook C, (2001). Brook’s Clinical Pediatric Endocrinology,
Blackwell publishing. With permission from Wiley Blackwell.
534 CHAPTER 7 Paediatric endocrinology
Further reading
Allen DB, Cuttler L, (2013). Clinical practice. Short stature in childhood - challenges and choices.
N Engl J Med 368, 1220–8.
Carel JC, Léger J (2008). Precocious puberty. N Engl J Med 358, 2366–77.
Clayton PE, Cianfarani S, Czernichow P, et aI. (2007). Management of the child born small for
gestational age child (SGA) through to adulthood: a consensus statement of the International
Societies of Paediatric Endocrinology and the Growth Hormone Research Society. J Clin
Endocrinology Metab Epub 2 January.
Clayton PE, Cuneo RC, Juul A, et al. (2005). Consensus statement on the management of the
GH-treated adolescent in the transition to adult care. Eur J Endocrinol 152, 165–70.
Conway GS (2009). Adult care of pediatric conditions: lessons from Turner’s syndrome. J Clin
Endocrinol Metab 94, 3185–7.
Dattani M, Preece M (2004). Growth hormone deficiency and related disorders: insights into causa-
tion, diagnosis, and treatment. Lancet 363, 1977–87.
De Waal WJ, Greyn-Fokker MH, Stijnen T, et al. (1996). Accuracy of final height prediction and
effect of growth-reductive therapy in 362 constitutionally tall children. J Clin Endocrinol Metab
81, 1206–16.
Growth Hormone Research Society (2000). Consensus guidelines for the diagnosis and treatment
of growth hormone (GH) deficiency in childhood and adolescence: summary statement of the
GH Research Society. GH Research Society. J Clin Endocrinol Metab 85, 3990–3.
Kelly WH, et al. (2012). Effect of inhaled glucocorticoids in childhood on adult height. N Engl J
Med 367, 904–12.
Saenger P, Wikland KA, Conway GS, et al. (2000). Recommendations for the diagnosis and manage-
ment of Turner’s syndrome. J Clin Endocrinol Metab 86, 3061–9.
536 CHAPTER 7 Paediatric endocrinology
Normal puberty
Puberty is the sequence of physical and physiological changes occurring at
adolescence, culminating in full sexual maturity.
Age at onset
Average age at onset of puberty is earlier in girls (711 years) than in boys
(712 years) but varies widely (792 years from the mean age of onset) and
is influenced by a number of factors:
• Historical. Age of menarche has d this century from 17 years in 1900
to 12.8 years today, presumably as a result of improved childhood
nutrition and growth.
• Genetic. Age at onset of puberty is partly familial.
• Ethnicity. Afro-Caribbean girls tend to have earlier puberty than
Caucasians.
• Weight gain. Earlier puberty is seen in girls who are overweight,
whereas girls who engage in strenuous activity and are thin often have
delayed puberty.
Hormonal changes prior to and during puberty
• Adrenal androgens (DHEAS and androstenedione) rise 2 years before
puberty starts (‘adrenarche’). This usually causes no physical changes
but occasionally results in early pubic hair and acne (‘premature
adrenarche’).
• Pulsatile secretion of LHRH from the hypothalamus at night is the first
step in the initiation of puberty and occurs well before physical signs
of puberty. This results in pulsatile secretion of LH and FSH from the
pituitary, and the gonadotrophin response to LHRH administration
reverses from the prepubertal FSH predominance to a higher response
in LH levels.
Physical changes
The first indication of puberty is breast development in girls and increase
in testicular size in boys. In each sex, puberty then progresses in an orderly
or ‘consonant’ manner through distinct stages (see Table 7.2). Puberty
rating by an experienced observer involves identification of pubertal
stage—particularly, breast development in girls and testicular volume (by
comparison with an orchidometer) in boys.
Pubertal growth spurt
i oestrogen levels in both boys and girls leads to i GH secretion. Peak
height velocity occurs at puberty stages 2–3 in girls and is later in boys at
stages 3–4 (testicular volume 10–12mL).
NORMAL PUBERTY 537
Precocious puberty
Definition
• Early onset of puberty is defined as <8 years in girls and <9 years in boys.
• Gonadotrophin-dependent (‘central’ or ‘true’) precocious puberty
is characterized by early breast development in girls or testicular
enlargement in boys.
• Gonadotrophin-independent puberty occurs due to abnormal
peripheral sex hormone secretion, resulting in isolated development
of certain s sexual characteristics. This may involve autonomous
testosterone production in a boy or autonomous oestrogen
production in a girl. In addition, testosterone production from the
adrenal or an ovarian tumour can induce virilization in a girl.
Assessment of precocious puberty
History
• Age when s sexual development first noted.
• What features are present and in what order did they appear? For
example, virilization (pubic, axillary, or facial hair; acne; body odour),
genital or breast enlargement, galactorrhoea (very rare), menarche, or
cyclical mood changes?
• Is there evidence of recent growth acceleration?
• Family history of early puberty?
• Past history of adoption or early weight gain or prior CNS abnormality
or insult (e.g. radiation)?
Examination
• Breast or genital and testicular size; degree of virilization
(clitoromegaly in girls indicates abnormal androgen levels).
• Neurological examination, particularly visual field assessment and
fundoscopy.
• Abdominal or testicular masses.
• Skin (? café-au-lait patches—McCune–Albright (b see Chapter 9,
McCune–Albright syndrome, pp. 576–7) or NF-1578).
• Assess height and height velocity over 4–6 months.
Investigations
The following investigations may be clinically indicated:
• Wrist radiograph for bone age.
• Thyroid function.
• Sex hormone levels (testosterone, oestrogen, androstenedione, DHEAS).
• LH and FSH levels (baseline and 30 and 60min post-IV LHRH).
• 17AOH progesterone levels (baseline and 30 and 60min post-IV
Synacthen®) if congenital adrenal hyperplasia suspected.
• Tumour markers (AFP, B-hCG).
• 24h urine steroid profile (see b p. 230).
• Abdominal US scan (adrenal glands, ovaries).
• MRI scan (cranial, adrenal glands).
• For those with hypogonadotrophic hypogonadism, mutation screening for
a monogenic cause (e.g. KA11, FGFRI, GnRHR, KISS1R) may be indicated
(see b p. 366).
PRECOCIOUS PUBERTY 539
McCune–Albright syndrome
(b also see McCune–Albright syndrome, pp. 576–7). This is a sporadic
condition due to a somatic activating mutation of the GSA protein subu-
nit which affects bones (polyostotic fibrous dysplasia), skin (café-au-lait
spots), and potentially multiple endocrinopathies.
A number of different hormone receptors share the same G
protein-coupled cyclic AMP second messenger system, and hyperthyroid-
ism or hyperparathyroidism may also be present.
All cells descended from the mutated embryonic cell line are affected
while cells descended from non-mutated cells develop into normal tissues.
Thus, the phenotype is highly variable in physical distribution and severity.
Testoxicosis
This is a rare familial condition, resulting in precocious puberty only in
boys due to an activating LH receptor mutation. Testes show only little
increase in size, and, on biopsy, Leydig cell hyperplasia is characteristic.
Treatment is by use of androgen receptor-blocking agents (cyproterone
acetate) and aromatase inhibitors.
Peripheral sex hormone secretion
• Excessive peripheral androgen secretion may occur due to CAH, or
androgen-secreting adrenal or gonadal tumours. These children usually
have rapid growth, advanced bone age, and moderate-to-severe
virilization in the absence of testicular or breast development.
(NB A testicular tumour may cause asymmetrical enlargement.)
• Peripheral oestrogen production from ovarian tumours is a rare cause
of precocious breast development in girls.
Premature thelarche
Premature breast development, in the absence of other signs of puberty,
may present at any age from infancy. Breast size may fluctuate and is often
asymmetrical. The cause is unknown, although typically FSH levels (but
not LH) are elevated, and ovarian US may reveal a single large cyst. Bone
maturation, growth rate, and final height are unaffected.
‘Thelarche variant’
This is an intermediate condition between premature thelarche and central
precocious puberty. The aetiology is unknown. These girls demonstrate i
height velocity and rate of bone maturation, and ovarian US reveals a
more multicystic appearance, as seen in true puberty. There is probably a
whole spectrum of presentations between premature thelarche and true
precocious puberty. Decision to treat should take into account height
velocity and final height prediction as well as the rate of physical matura-
tion and the severity of accompanying pubertal features (e.g. mood swings
and difficult behaviour).
PRECOCIOUS PUBERTY 541
Delayed/absent puberty
Definition
Delayed puberty is defined as failure to progress into puberty by >2
SDs later than the average, i.e. >13 years in girls and >14 years in boys.
Clinically, boys are more likely to present with delayed puberty than girls.
In addition, some children present with delay in progression from one
pubertal stage to the next for >2 years.
Psychological distress may be exacerbated by declining growth velocity
relative to their peers. In the long term, severe delay may be a risk factor
for d bone mineral density and osteoporosis.
Causes
General
• Constitutional delay of growth and puberty (this is the most common
cause; b see Constitutional delay of growth and puberty, p. 520).
• Chronic childhood disease, malabsorption (e.g. coeliac disease,
inflammatory bowel disease), or undernutrition.
Hypergonadotrophic hypogonadism
Gonadal failure may be:
• Congenital (e.g. Turner’s syndrome in girls (b p. 336), Klinefelter’s
syndrome in boys (b p. 370)).
• Acquired (e.g. following chemotherapy, local radiotherapy, infection,
torsion).
In these conditions, basal and stimulated gonadotrophin levels are raised.
Gonadotrophin deficiency
• Kallman’s syndrome (b p. 362) (including anosmia).
• HP lesions (tumours, post-radiotherapy, dysplasia).
• Rare inactivating mutations of genes encoding LH, FSH (or their
receptors).
These conditions may also present in the newborn period with micropenis
and undescended testes in boys.
Investigation
The following investigations may be clinically indicated:
• LH and FSH levels (basal and post-IV LHRH stimulation).
• Plasma oestrogen or testosterone levels.
• Measurement of androgen levels before and after hCG therapy may be
used to indicate presence of functional testicular tissue in boys.
• Karyotype.
• Pelvic US in girls to determine ovarian morphology.
• US or MRI imaging in boys to detect intra-abdominal testes.
• For those with hypogonadotropic hypogonadism, mutation screening for
a monogenic cause (e.g. KALI, FGFRI, GnRHR, KISSIR) may be indicated.
It may be difficult to distinguish between constitutional delay and gonadotro-
phin deficiency, as gonadotrophin levels are low in both conditions. In these
cases, induction of puberty may be indicated, with regular assessment of testi-
cular growth in boys (which is independent of testosterone therapy), followed
by withdrawal of treatment and reassessment when final height is reached.
DELAYED/ABSENT PUBERTY 543
Management
Depending on the age and concern of the child and parents, short-course
exogenous sex steroids can be used to induce pubertal changes. If gonado-
trophin deficiency is permanent or the gonads are dysfunctional or absent,
then exogenous sex steroids are required to induce and maintain pubertal
development (b see Constitutional delay of growth and puberty, p. 520).
Long-term treatment
(See Table 7.3.)
Boys
Testosterone (by IM injection) 50mg 4–6-weekly, gradually i to 250mg
4-weekly.
• Monitor penis enlargement, pubic hair, height velocity, and adult body
habitus.
• Side effects include severe acne and, rarely, priapism.
Girls
Oestrogen (oral). Start at low dose (ethinylestradiol 2 micrograms daily),
and gradually increase.
• Promotes breast development and adult body habitus.
• Progesterone (oral) should be added if breakthrough bleeding occurs or
when oestrogen dose reaches 10 micrograms/day.
Further reading
Hindmarsh PC (2009). How do you initiate oestrogen therapy in a girl who has not undergone
puberty? Clin Endocrinol (Oxf) 71, 7–10.
Miller JF (2012). Approach to the child with Prader-Willi syndrome. J Clin Endocrinol Metab 97,
3837–44.
Palmert MR, Dunkel L (2012). Clinical practice. Delayed puberty. N Engl J Med 366, 443–53.
544 CHAPTER 7 Paediatric endocrinology
Gonadotrophin defects
• Gonadotrophin deficiency may occur in hypopituitarism or may be
associated with anosmia (Kallmann’s syndrome). It usually presents
with delayed puberty but is an occasional cause of micropenis and
undescended testes.
• LH receptor gene defects are rare and result in complete absence of
virilization, as the testes are unable to respond to placental hCG.
Anti-Müllerian hormone deficiency or insensitivity
Testes are usually undescended, and uterus and Fallopian tubes present.
Management of ambiguous genitalia
In the newborn period, the infant should be monitored in hospital for:
• Hypoglycaemia (until hypopituitarism is excluded).
• Salt wasting (until CAH is excluded).
Explain to the parents that the infant appears to be healthy but has a
defect that interferes with determining sex. It is helpful to show the par-
ents the physical findings as you explain this. Advise them to postpone
the registration of the birth until after further investigations, and discuss
what they will say to relatives and friends. The sex of the baby should be
assigned as soon after birth as is practicable, given the need for accurate
diagnosis.
548 CHAPTER 7 Paediatric endocrinology
Sex assignment
Decision on the sex of rearing should be based on the optimal expected
outcome in terms of psychosexual and reproductive function. Parents
should, therefore, be encouraged discussion with an endocrinologist,
a surgeon specializing in urogenital reconstruction, a psychologist, and a
social worker. Following the necessary investigations and discussions, early
gender assignment optimizes the psychosexual outcome.
Further management
• If ♀ sex is assigned, any testicular tissue should be removed.
• Reconstructive surgery may include clitoral reduction, gonadectomy,
and vaginoplasty in girls, and phallus enlargement, hypospadias repair,
and orchidopexy in boys. In both sexes, multiple-stage procedures may
be required.
• Topical or systemic dihydrotestosterone may enhance phallus size
in ♂ infants, and a trial of therapy is sometimes useful before sex
assignment.
• Hormone replacement therapy may also be required from puberty
into adulthood.
• Continuing psychological support for the parents and children is very
important.
DISORDERS OF SEX DEVELOPMENT (DSD) 549
550 CHAPTER 7 Paediatric endocrinology
• Plasma and urine electrolytes may need to be monitored over the first
2 weeks.
• Plasma renin level is also useful to confirm salt wasting in those with
normal serum sodium and relatively mild 21-hydroxylase deficiency.
Other rare enzyme deficiencies
• 17A-hydroxylase deficiency. Impairs cortisol, androgen, and oestrogen
synthesis, but overproduction of mineralocorticoids leads to
hypokalaemia and hypertension.
• 3B-hydroxysteroid dehydrogenase deficiency. Impairs cortisol,
mineralocorticoid, and androgen biosynthesis. ♂ have hypospadias
and undescended testes; however, excess DHEA, a weak androgen,
may cause mild virilization in ♀.
Steroid acute regulatory protein
• STAR mutation: 46XY sex reversal and severe adrenal failure.
Heterozygous defect. Extremely rare.
• CYPIIAI (P450: side chain cleavage cytochrome enzyme): 46XY sex
reversal and severe adrenal failure. Heterozygous defect. Extremely rare.
• P450 oxidoreductase deficiency: biochemical picture of combined
21-hydroxylase/17A-hydroxylase/17,20 lyase deficiencies. Spectrum of
presentation from children with ambiguous genitalia, adrenal failure,
and the Antley–Bixler skeletal dysplasia syndrome through to mildly
affected individuals with polycystic ovary syndrome.
Management
• Hydrocortisone (15mg/m2 per day orally in three divided doses).
In addition to treating cortisol deficiency, this therapy suppresses
ACTH and thereby limits excessive production of adrenal androgens.
Occasionally, higher doses are required to achieve adequate androgen
suppression; however, overtreatment may suppress growth.
• In salt losers, initial IV fluid resuscitation (10–20mL/kg normal
saline) may be required to treat circulatory collapse. Long-term
mineralocorticoid replacement (fludrocortisone 0.05–0.3mg/day) may
have incomplete efficacy, particularly in infancy, and sodium chloride
supplements are also needed. Up to 10mmol/kg per day may be
needed in infancy.
• Reconstructive surgery (clitoral reduction, vaginoplasty). Often
performed in infancy, and further procedures may be required during
puberty. Needs experienced surgeon.
• Patients and families need to be aware of the need for extra
hydrocortisone during intercurrent illness or significant stress.
Instructions on what to do for mild, moderate, and severe illnesses
(‘Sick Day Rules’) and for operative procedures should be made
available (see b Addison disease treatment, p. 266). Patients and
families should have parenteral hydrocortisone available to give in
emergency situations.
552 CHAPTER 7 Paediatric endocrinology
Neuroendocrine disorders
Classification of neuroendocrine
neoplasias (NENs)
• There is a move towards standardizing the terminology of
these tumours; the European Neuroendocrine Society (ENETS)
recommends the use of the umbrella term neuroendocrine neoplasia
(NEN). The term NEN includes low- and intermediate-grade neoplasia
(previously referred to as carcinoid or atypical carcinoid) which are
now referred to as neuroendocrine tumours (NETs) and high-grade
neoplasia (neuroendocrine carcinoma, NEC). There is a confusing
array of classifications of NENs, based on anatomical origin, histology,
and secretory activity.
• Many of these classifications are well established and widely used.
• The WHO classifications of lung and thymic NETs and
gastroenteropancreatic (GEP) NETs are shown in Table 8.1.
• The name carcinoid continues to be used in the WHO classification of
lung and thymic NETs. See Table 8.1.
• It is important to understand the differences between ‘differentiation’,
which is the extent to which the neoplastic cells resemble their
non-tumourous counterparts, and ‘grade’, which is the inherent
aggressiveness of the tumour.
• The grades (low, intermediate, or high grade) in lung and thymic
NETs are based on mitoses and necrosis. In GEP NETs, in addition
to mitoses, the Ki67 index (a marker of proliferation) determines
the grade.
• Neuroendocrine carcinomas are the most aggressive NENs and can
either be small or large cell type.
Clinical features of NENs
NENs are diagnosed based on histological features of biopsy specimens.
The presenting features of the tumours vary like any other tumour, based
on their anatomical location, such as abdominal pain, intestinal obstruc-
tion. Many are incidentally discovered during endoscopy or imaging for
unrelated conditions. In a database study, 49% of NENs were localized,
24% had regional metastases, and 27% had distant metastases. The NENs
most associated with metastases are pancreatic, followed by caecal,
colonic, and small intestine in reducing frequency. These tumours rarely
manifest themselves due to their secretory effects.
Treatment of NENs
A multidisciplinary approach to the treatment of NENs is essential.
Surgical treatment
• Surgery is the treatment of choice for NENs grades 1 and 2, except
in the presence of widespread distant metastases and extensive local
invasion.
• Preoperative octreotide may prevent carcinoid crisis.
• In patients with widespread local disease or distant metastases, surgery
could still be beneficial by reducing tumour bulk. When debulking
liver metastases, intraoperative ablation may be used as adjunctive
treatment. This is helpful in symptom control.
• Surgery may be required to alleviate intestinal obstruction.
• There are surgical standards (ENETS) for the treatment of GEP NENs.
• There are limited data to set standards for the surgical management of
bronchial and extra-gastrointestinal tumours.
Ablative therapies
• For NENs with liver metastases, radiofrequency ablation or
transarterial chemoembolization are used.
Medical management
• The endpoints of clinical trials are either progression-free survival or
time to progression due to their slow growth. This is a fundamental
difference compared with adenocarcinomas.
• For neuroendocrine carcinomas, chemotherapeutic options are similar
to those for small cell lung cancer.
• Medical management for symptom control and disease progression is
applicable when potentially curative surgery and ablative procedures
are not possible.
See Box 8.1 for non-functioning pancreatic NENs.
Medical therapies that are available are:
Somatostatin analogues
• Somatostatin analogues (SSA) have relatively minor side effects and
provide long-term symptom control.
• Octreotide and lanreotide and their longer-acting analogues reduce
the level of biochemical tumour markers in the majority of patients
and control symptoms in around 70% of cases.
• SSAs may be beneficial in functioning and non-functioning metastatic
small intestinal G1 NET for tumour stabilization. They are not
recommended for use in metastatic G3 NEC.
• Radiolabelled somatostatin analogues with yttrium-90 alleviate
symptoms of metastatic tumours and may improve quality of life.
• Pasireotide, a newer somatostatin analogue which has a high binding
affinity to four out of the five human somatostatin receptor subtypes,
has a longer half-life. This drug is being investigated for use in NENs in
conjunction with newer molecular targeted therapies.
TREATMENT OF NENS 559
Interferon α
• There is reasonable biochemical response (about 45%), with
improvement in symptoms.
• A combination of interferon with octreotide has been shown to
produce biochemical and symptomatic improvement in patients who
have previously had no significant benefit from either drug alone.
Chemotherapy
Cytotoxic chemotherapy may be considered in patients with progressive,
advanced, or uncontrolled symptomatic disease. It is recommended in
metastatic pancreatic NET, foregut metastatic G2 NET, and NEC G3 of
any primary. It is not recommended in well-differentiated midgut NET.
• Streptozotocin-based combinations, such as with fluorouracil, have
been historically in use. Three-drug regimens, including cisplatin or
doxorubicin, show superior response rates compared to the two-drug
regimen.
• Capecitabine (prodrug of 5FU) is administered orally and may offer
more antitumour effect.
• Like streptozotocin, dacarbazine is an alkylating agent and has shown
to be of benefit in the treatment of pancreatic NETs.
• Temozolomide, another alkylating agent and an orally administered
analogue of dacarbazine, is associated with less toxicity. However, data
are limited.
Molecular therapies
These therapies target signalling pathways, such as tyrosine kinase (TK)
and mammalian target of rapamycin (sirolimus) (mTOR), which have a
regulatory role in neuroendocrine cell growth.
• The small molecule TK inhibitor sunitinib has been shown to
significantly increase progression-free survival in patients with
malignant pancreatic NETs when compared with placebo.
• The mTOR inhibitor everolimus has been shown to reduce the risk of
progression by 65% vs placebo in a recent study. It may also be used
with octreotide.
560 CHAPTER 8 Neuroendocrine disorders
Further reading
European Neuroendocrine Tumour Society. Available at: M http://www.enets.org.
Pavel ME, et al. (2011). Everolimus plus octreotide long-acting repeatable for the treatment of
advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a ran-
domised, placebo-controlled, phase 3 study. Lancet 378, 2005–12.
Raymond E, et al. (2011). Sunitinib malate for the treatment of pancreatic neuroendocrine tumors.
N Engl J Med 364, 501–13.
Yao JC, et al. (2011). Everolimus for advanced pancreatic neuroendocrine tumors. N Engl J Med
364, 514–23.
562 CHAPTER 8 Neuroendocrine disorders
Insulinomas
Definitions and background
• An insulinoma is a functioning neuroendocrine tumour of the pancreas
that causes hypoglycaemia through inappropriate secretion of insulin.
• Unlike other neuroendocrine tumours of the pancreas, more than 90%
of insulinomas are benign.
• >80% of insulinomas are solitary, but some are multiple, either
simultaneously or consecutively, a situation likely to be associated with
MEN-1 in approximately 10%.
• Insulinomas are found with equal frequency throughout the head,
body, and tail of the pancreas.
Epidemiology
• The annual incidence of insulinomas is of the order of 1–2 per million
population.
• There is a slight female preponderance in the fifth decade of life.
• 5–10% of insulinomas are associated with MEN-1.
Clinical presentation
• Patients may present with neuroglycopenic symptoms, including
headaches, diplopia, blurred vision, altered behaviour, and sometimes
seizures.
• Whipple’s triad remains as useful as ever in diagnosing
insulinoma: symptoms of hypoglycaemia, plasma glucose (laboratory
glucose—and not fingerprick readings) of 2.2mmol/L or less, and
resolution of symptoms with glucose.
Biochemical investigations
• The gold standard investigation for establishing the diagnosis of
insulinomas is the 72h fast.
• Shorter durations of fast (15h), repeated on three separate occasions,
may be used as a screening tool.
Diagnostic criteria
• Laboratory blood glucose of ≤2.2mmol/L.
• Concomitant inappropriate insulin elevation.
• Elevated C-peptide and proinsulin.
• β-hydroxybutyrate of <2.7mmol/L.
• Absence of sulfonylureas and metabolites in plasma or urine.
TUMOUR LOCALIZATION 563
Tumour localization
(See Table 8.2.)
• Islet cell tumours are often small and may not be detected by any
imaging technique. The available radiological modalities have a wide
reported range of sensitivity which is frequently dependent on the
equipment and the operator.
• MRI or spiral CT correctly detects >60% of tumours, but preoperative
localization can be difficult in tumours <1cm in diameter.
• Arterial stimulation (with calcium or secretin), followed by venous
sampling, can localize approximately 90% of insulinomas but carries the
risks of a more invasive technique.
• Endoscopic US, which requires specialized equipment and expertise,
may identify tumours as small as 5mm. The head of the pancreas is
visualized with the probe in the duodenum, and the body and tail with
it in the stomach.
• Intraoperative US, using a transducer applied directly to the pancreas,
improves the sensitivity of US to 790%.
• Experienced surgeons can frequently identify the lesions
intraoperatively by palpation alone, and the risk of multiple tumours
makes a thorough examination of the whole pancreas essential at
operation.
• Somatostatin receptor scintigraphy has been shown to be useful in
detecting insulinomas, but it is dependent on somatostatin receptor
subtype expression by the tumour.
• Glucagon-like peptide-1 receptor imaging has also been shown to be
useful.
Treatment of insulinomas
• The treatment of choice in all, but poor, surgical candidates is
operative removal.
• Surgical options include enucleation of tumours, partial
pancreatectomy, Whipple procedure, or rarely total pancreatectomy.
• In experienced surgical hands, the mortality is less than 1%. The
mortality is largely influenced by the incidence of acute post-operative
pancreatitis and peritonitis.
• Post-operative hyperglycaemia may occur, even following partial
pancreatectomy.
• Medical treatment to control symptoms may be achieved using
diazoxide alone or in combination with octreotide pending surgery or
in those patients who have high surgical risk.
• Radiolabelled somatostatin analogue therapy may induce a biochemical
and symptomatic response in patients with malignant insulinomas.
• Everolimus, the M Tor inhibitor, may be useful in metastatic insulinoma
but has significant side-effects.
Prognosis of insulinomas
• Following the removal of a solitary insulinoma, life expectancy is
restored to normal.
• Malignant insulinomas, with metastases usually to the liver, have a
natural history of years, rather than months, and may be controlled
with medical therapy or specific antitumour therapy using
streptozotocin and fluorouracil. The management goals are targeted at
managing blood glucose and tumour load.
• Average 5-year survival estimated to be approximately 35% for
malignant insulinomas.
Further reading
Carl Pallais J, et al. (2012). Case records of the Massachusetts General Hospital. Case 33-2012.
A 34-year-old woman with episodic paresthesias and altered mental status after childbirth. N
Engl J Med 367, 1637–46.
de Herder WW, et al. (2011). New therapeutic options for metastatic malignant insulinomas. Clin
Endocrinol (Oxf) 75, 277–84.
Placzhowski KA, et al. (2009). Secular trends in the presentation and management of functioning
insulinoma at the Mayo Clinic, 1987-2007. J Clin Endocrinol Metab 94, 1069–73.
GASTRINOMAS 565
Gastrinomas
Definitions
• Gastrin, synthesized in the G cells, predominantly in the gastric
antrum, is the principal gut hormone stimulating gastric acid secretion.
• The Zollinger–Ellison (ZE) syndrome is characterized by gastric acid
oversecretion and manifests itself as severe peptic ulcer disease
(PUD), gastro-oesophageal reflux, and diarrhoea.
Incidence
The incidence of gastrinomas is 0.5–2/million population/year.
Sites of origin
• Gastrinomas are the most common functional malignant pancreatic
endocrine tumours.
• The most common extrapancreatic site of origin is the duodenum in
50–80% of sporadic ZE and 70–100% of patients with MEN-1.
• Less common sites are stomach, liver, bile duct, heart, and small cell
lung cancer.
• Pancreatic tumours are more aggressive than duodenal tumours.
Clinical presentation
• The mean age of presentation of patients with sporadic gastrinomas is
48–55 years and is almost equally found in ♂ and ♀.
• Most patients with ZE syndrome present with a single duodenal ulcer
or gastro-oesophageal reflux.
• Abdominal pain is a presenting feature (due to PUD) in 75–98% of
patients.
• Diarrhoea is present at diagnosis in 30–70%.
• 20–30% of patients with gastrinomas have MEN-1.
• At diagnosis, 5–10% of duodenal gastrinomas and 20–25% of pancreatic
gastrinomas have hepatic metastases.
• Suspect ZE syndrome when there is severe PUD without Helicobacter
pylori, resistance to PPIs, or in the presence of hypercalcaemia.
566 CHAPTER 8 Neuroendocrine disorders
Treatment of gastrinomas
(See Table 8.3.)
• The treatment of choice is complete surgical tumour removal,
although this is usually only considered after the tumour has been
identified preoperatively.
• Surgery is rarely justified in patients with known hepatic metastases,
although some small studies have raised the possibility of benefit from
tumour debulking.
• Where preoperative imaging has failed to identify a tumour, the
patient is often best maintained on high-dose PPI therapy (e.g. 60mg of
omeprazole), with regular imaging to reassess. Check B12 annually.
• Somatostatin analogues do not appear to be more effective at
controlling gastric acid hypersecretion and relieving symptoms than
PPIs or H2 blockers.
• The data are currently few, but, as for insulinomas, radiolabelled
somatostatin analogues may be a future treatment option for
gastrinomas.
• In patients with MEN-1, management is more controversial, but
the usual policy is to operate when a well-defined tumour can be
identified.
Prognosis of gastrinomas
• 10-year survival without liver metastases is 95%.
• 10-year survival with single or limited metastases in both lobes is
approximately 80%.
• Where there are diffuse metastases, the outlook is less favourable,
with a 10-year survival of approximately 15%.
Further reading
Simmons LH, et al. (2013). Case records of the Massachusetts General Hospital. Case 6-2013.
A 54-year-old man with recurrent diarrhea. N Engl J Med 368, 757–65.
568 CHAPTER 8 Neuroendocrine disorders
Glucagonomas
Definitions
• Glucagonomas are neuroendocrine tumours that usually arise from
the A cells of the pancreas and produce the glucagonoma syndrome
through the secretion of glucagon and other peptides derived from the
preproglucagon gene.
• The large majority of glucagonomas are malignant, but they are also very
indolent tumours, and the diagnosis may be overlooked for many years.
• Up to 90% of patients will have lymph node or liver metastases at the
time of presentation.
• They are classically associated with the rash of necrolytic migratory
erythema.
Epidemiology
• The annual incidence is estimated at 1 per 20 million population.
• Glucagonomas are uncommon in MEN-1.
Clinical presentation
(See Table 8.4.)
• The characteristic rash—necrolytic migratory erythema—occurs in
>70% of cases and usually manifests initially as a well-demarcated area
of erythema in the groin before migrating to the limbs, buttocks, and
perineum.
• Mucous membrane involvement is common, with stomatitis, glossitis,
vaginitis, and urethritis being frequent features.
• Glucagon antagonizes the effects of insulin, particularly on hepatic
glucose metabolism, and glucose intolerance is a frequent association
(>90%).
• Sustained gluconeogenesis also causes amino acid deficiencies and
results in protein catabolism which can be associated with unrelenting
weight loss in >60% of patients.
• Glucagon has a direct suppressive effect on the bone marrow, resulting
in a normochromic normocytic anaemia in almost all patients.
Biochemical investigations
• The diagnosis is confirmed on finding raised plasma glucagon levels.
• A gut hormone profile may also show elevated neuroendocrine
markers, such as pancreatic polypeptide, chromogranin A.
• Impaired glucose intolerance and hypoaminoacidaemia may be present.
Tumour localization
• At the time of diagnosis, 50–100% of glucagonomas will have
metastasized to the liver, and most p tumours will be >3cm in
diameter.
• These tumours rarely present problems of radiological localization,
and transabdominal US and CT scanning are usually adequate.
• Small tumours may require more sophisticated imaging techniques,
including endoscopic ultrasound (see Table 8.4). Octreotide scanning
probably offers the best means of evaluating the extent of metastatic
disease.
GLUCAGONOMAS 569
Treatment
• Surgery is the only curative therapeutic option, but the potential for a
complete cure may be as low as 5%.
• Somatostatin (SST) analogues are the treatment of choice, with
excellent response rates in treating the necrolytic migratory erythema.
They are less effective at reversing the weight loss and have an
inconsistent effect on glycaemic control such that diabetes mellitus
may need to be managed with insulin therapy.
• If SST analogues fail, the rash may be improved using IV amino acids
and fatty acids.
• Experience with chemotherapy is limited. However, palliative
chemotherapy, using streptozotocin and fluorouracil, has been shown
to produce a 50% reduction in glucagon levels in 75% of patients, but
the benefit is frequently only temporary. Antitumour activity has also
been shown with the alkylating agent temozolomide.
• Hepatic artery embolization may result in a dramatic relief of
symptoms, with remissions of several months recorded.
• As for the other pancreatic islet cell tumours, there are data to suggest
that radiolabelled somatostatin analogues may be therapeutic in the
glucagonoma syndrome.
• Nutritional support is essential, as patients may experience a
prolonged catabolic state.
570 CHAPTER 8 Neuroendocrine disorders
VIPomas
Definitions
• In 1958, Verner and Morrison1 first described a syndrome consisting
of refractory watery diarrhoea and hypokalaemia, associated with a
neuroendocrine tumour of the pancreas.
• The syndrome of watery diarrhoea, hypokalaemia, and acidosis
(WDHA) is due to secretion of vasoactive intestinal polypeptide (VIP).
• Tumours that secrete VIP are known as VIPomas.
• VIPomas account for <10% of islet cell tumours and mainly occur as
solitary tumours.
• >60% are malignant and metastasize to the lymph nodes, liver, kidneys,
and bone.
Clinical presentation
• The most prominent symptom in most patients is profuse watery
diarrhoea which is secretory in nature and, therefore, rich in
electrolytes (see Box 8.3).
• Other causes of secretory diarrhoea should be considered in the
differential diagnosis (see Box 8.4).
• VIPomas are rare in MEN-1 patients, occurring in <1% of cases.
Biochemical investigations
• Elevated levels of plasma VIP are found in all patients with the VIPoma
syndrome, although false +ves may occur in dehydrated patients due
to diarrhoea from other causes.
• A gut hormone profile may identify other raised tumour markers, such
as pancreatic polypeptide, and aid detection of some other causes of
watery diarrhoea, e.g. gastrinomas.
• A phaeochromocytoma screen should be performed, especially in
children in whom it is common to find the tumours residing in the
adrenal medulla.
Tumour localization
• The majority of tumours secreting VIP originate in the pancreas while
others arise from the sympathetic chain.
• p VIPomas have very rarely been reported to arise from a variety
of other sites, such as the lung, oesophagus, small bowel, colon, and
kidney.
• Most patients present with large tumours which can be easily identified
by transabdominal US or CT, although small tumours may require
additional methods of tumour localization, as previously discussed (see
Table 8.2).
Treatment
• Severe cases require IV fluid replacement and careful correction of
electrolyte disturbances.
• Surgery to remove the tumour is the treatment of first choice, if
technically possible, and may be curative in around 40% of patients.
Surgical debulking may also be of palliative benefit.
• Somatostatin analogues produce effective symptomatic relief from the
diarrhoea in most patients. Long-term use does not result in tumour
regression.
• Glucocorticoids in high dosage have also been shown to provide good
relief of symptoms.
• A trial of lithium may be warranted in resistant cases, and this therapy
may be combined with octreotide.
• Chemotherapy using streptozotocin, in combination with fluorouracil,
has resulted in response rates of >30%.
• Hepatic artery embolization can offer temporary respite from severe
diarrhoea.
Reference
1. Verner JV, Morrison AB (1958) Islet cell tumor and a syndrome of refractory watery diarrhea
and hypokalemia. Am J Med 25, 374–380.
572 CHAPTER 8 Neuroendocrine disorders
Somatostatinomas
Definitions
• Somatostatinomas are very rare neuroendocrine tumours, occurring
both in the pancreas and in the duodenum.
• >60% are large tumours located in the head or body of the pancreas.
• The clinical syndrome may be diagnosed late in the course of the
disease when metastatic spread to local lymph nodes and the liver has
already occurred.
Clinical features
(See Box 8.5.)
• Glucose intolerance or frank diabetes mellitus may have been
observed for many years prior to the diagnosis and retrospectively
often represents the first clinical sign. It is probably due to the
inhibitory effect of somatostatin on insulin secretion.
• A high incidence of gallstones has been described similar to that seen
as a side effect with long-term somatostatin analogue therapy.
• Diarrhoea, steatorrhoea, and weight loss appear to be consistent
clinical features and may be associated with inhibition of the exocrine
pancreas by somatostatin.
• Small duodenal somatostatinomas may occur in association with NF-1,
and, although these rarely cause the inhibitory clinical syndrome, they
present with obstructive biliary disease through local spread of the
tumour.
• Somatostatinomas are infrequently associated with MEN-1 (7%).
Biochemical investigations
• Plasma somatostatin levels will be raised and may also be associated
with raised levels of other neuroendocrine tumour markers, including
ACTH and calcitonin.
• Multisecretory activity is commoner with pancreatic (33%) than with
duodenal (16%) somatostatinomas.
Tumour localization
• Transabdominal US and CT scanning may demonstrate the tumour
since metastatic disease is often apparent at presentation.
• Additonal methods of tumour localization, as previously described,
may also be required (see Table 8.2).
Treatment
• Surgery should be considered as first-line treatment as, although a
cure is rare, even debulking surgery may result in significant palliation.
• Hepatic embolization can be considered, and chemotherapy with
streptozotocin and fluorouracil may be used to control malignant
disease.
SOMATOSTATINOMAS 573
Inherited endocrine
syndromes and MEN
McCune–Albright syndrome
Definitions
The syndrome is characterized by:
• Polyostotic fibrous dysplasia.
• Café-au-lait pigmented skin lesions.
• Autonomous function of multiple endocrine glands.
A clinical diagnosis requires two of these pathologies.
Epidemiology
The condition affects 1:100,000 to 1:1,000,000 people.
Genetics
• A genetic, but not an inherited, condition due to a post-zygotic
somatic mutation in the gene (GNAS1) that encodes the A-chain of the
stimulating G protein of adenyl cyclase (GSA mutation). This results in
activation of adenyl cyclase.
• The somatic mutation results in mosaicism, and consequently the
proportion and distribution of affected cells in a tissue will be
determined by the precise stage in development at which the mutation
occurred.
• Mutational analysis of the GNAS1 gene from affected tissues or blood
is available in some centres in the UK.
Clinical features
Polyostotic fibrous dysplasia
• Solitary or multiple expansile bony lesions which can cause fracture
deformities and nerve entrapment typically develop before the age of
10 years.
• The femora and the pelvic bones are most frequently affected, and
radiographs of these bones are useful for screening.
• Osteosarcomas are a rare complication.
• For treatment, see Box 9.1.
Café-au-lait pigmentation
The lesions are characterized by an irregular border (in neurofibroma-
tosis, the border is smooth), do not cross the midline, and tend to be
ipsilateral to the bone lesions.
Endocrinopathies
See Table 9.1.
Involvement of other organs
• Hepatobiliary complications, such as neonatal jaundice, elevated
transaminases, and cholestasis are relatively common.
• Cardiomegaly, tachyarrhythmias, and sudden cardiac death may occur.
• Gastrointestinal polyps, splenic hyperplasia, and pancreatitis are
reported complications.
• Recognized CNS associations include microcephaly, failure to thrive,
and developmental delay.
MCCUNE–ALBRIGHT SYNDROME 577
Prognosis
• Most patients live well beyond reproductive age.
• Bone deformities may reduce life expectancy.
• Sudden cardiac death, although recognized, is uncommon.
Further reading
Lumbroso S, Paris F, Sultan C (2002). McCune–Albright syndrome: molecular genetics. J Paediatr
Endocrinol Metab 15, 875–82.
Spiegel AM, Weinstein LS (2004). Inherited diseases involving G proteins and G protein-coupled
receptors. Annu Rev Med 55, 27–39.
578 CHAPTER 9 Inherited endocrine syndromes
Neurofibromatosis
Definitions
• Neurofibromatosis type 1 (NF1) is also known as von Recklinghausen
disease and refers to the occurrence of multiple neurofibromas,
café-au-lait spots, and Lisch nodules (pigmented hamartomas nodules
of aggregated dendritic melanocytes) affecting the iris.
• Endocrinopathies are sometimes associated with NF1.
• NF type 2 (NF2) is characterized by the presence of bilateral acoustic
neuromas, typically resulting in deafness.
• Other features of NF2 include posterior subcapsular cataracts, retinal
gliomas, pigmented retinopathy, and gaze palsies.
• NF2 has no common associated endocrinopathies.
• NF2 is rare, with an estimated frequency of 1 in 40,000 live births.
NF1
Genetics
• NF1 is a highly penetrant, autosomal dominant condition.
• The NF1 gene is located on chromosome 17 and encodes a
GTPase-activating protein (neurofibromin).
• Neurofibromin promotes cleavage of GTP to GDP.
Epidemiology
• The incidence of NF1 is 1 per 3,000 of the population.
• NF1 is nearly 100% penetrant but shows variable clinical expression.
• Approximately 50% of cases are sporadic.
Clinical features
• Diagnosis is generally apparent by the age of 1 year.
• The café-au-lait spots become visible shortly after birth (95%); 70%
have axillary or groin freckling.
• The multiple cutaneous and subcutaneous neurofibromas appear
around puberty (95%).
• Lisch nodules, affecting the iris, start to appear typically after the age of
5 years (95%).
• The endocrine features are detailed in Box 9.2.
• Learning disabilities occur in 60% of patients with NF1.
• Gliomas may be found in around 15% of patients, most commonly
affecting the optic pathways.
• Neurofibrosarcomas complicate around 6% of cases.
• Skeletal dysplasias (e.g. sphenoid wing dysplasia, scoliosis, tibial
pseudoarthrosis, pectus excavatum, etc.) may be found in up to 5%
of cases.
• Vascular dysplasia may occur, with the commonest region affected
being the renal vasculature, resulting in renovascular hypertension in
up to 3% of cases.
• Macrocephaly (16%), seizures (5%), and short stature (6%) are all
reported features of NF1.
• Associated phaeochromocytomas are seen in <5% of NF1 patients.
NEUROFIBROMATOSIS 579
Further reading
Arun D, Gutmann DH (2004). Recent advances in neurofibromatosis type 1. Curr Opin Neurol
17, 101–5.
Rose VM (2004). Neurocutaneous syndromes. Mo Med 101, 112–16.
580 CHAPTER 9 Inherited endocrine syndromes
Clinical features
• Retinal angiomas are the initial manifestation of VHL in 40% of patients.
They are uncommon before the age of 10 years but continue to
develop throughout life. They tend to be peripheral in the retina,
appearing as red oval lesions. Bleeding and retinal detachment may
occur, and treatment is with laser therapy.
• 75% of haemangioblastomas occur in the cerebellum, and they are the
initial presenting feature in 40% of VHL patients. Treatment is with
surgery or radiotherapy.
• Renal carcinoma is the commonest cause of death in VHL patients. By
the age of 60 years, 70% of VHL patients will be affected, with a mean
age of presentation of 44 years. The lesions tend to be multifocal. The
management of choice is surgical resection.
582 CHAPTER 9 Inherited endocrine syndromes
Carney complex
Definitions
A clinical diagnosis is made by finding two of the clinical features listed in
Clinical presentation below or one of these features plus either an affected
first-degree relative or an inactivating mutation in the gene PRKARIα.
Genetics
• Autosomal dominant.
• An inactivating mutation of the PRKARIA gene on the long arm of
chromosome 17q2 can be identified in approximately 50% of families.
Clinical presentation
• Spotty skin pigmentation.
• Cardiac, skin, or mucosal myxomas.
• Endocrine tumours (see Box 9.4).
• Psammomatous melanotic schwannoma.
Further reading
Bertherat J, et al. (2009). Mutations in regulatory subunit type 1A of cyclic adenosine
5'-monophosphate-dependent protein kinase (PRKAR1A): phenotype analysis in 353 patients
and 80 different genotypes. J Clin Endocrinol Metab 94, 2085–91.
Sandrini F, Stratakis C (2003). Clinical and molecular genetics of Carney complex. Mol Genet Metab
78, 83–92.
Stergiopolous SG, Abu-Asab MS, Tsokos M, et al. (2004). Pituitary pathology in Carney complex
patients. Pituitary 7, 73–82.
Stratakis CA, Kirschner LS, Carney JA (2001). Clinical and molecular features of the Carney com-
plex. J Clin Endocrinol Metab 86, 4041–6.
584 CHAPTER 9 Inherited endocrine syndromes
Cowden syndrome
Clinical presentation
(Also see Box 9.5.)
• The condition is characterized by multiple hamartomas, involving organ
systems derived from all three germ cell layers.
• Patients are also at risk from breast, thyroid, and endometrial
carcinomas.
• Thyroid pathology occurs in >75% of patients, and the lesions may be
multifocal.
• Weight gain is common.
Epidemiology
Estimated to affect 1 in 200,000 individuals.
Genetics
• Autosomal dominant condition.
• Inactivating mutations in the PTEN tumour suppressor gene can be
identified in approximately 80% of affected probands.
Further reading
Pilarski R, Eng C (2004). Will the real Cowden syndrome please stand up (again)? Expanding muta-
tional and clinical spectra of the PTEN hamartoma tumour syndrome. J Med Genet 41, 323–6.
POEMS SYNDROME 585
POEMS syndrome
Definitions
Progressive polyneuropathy, organomegaly, endocrinopathy, monoclo-
nal gammopathy, and skin changes (POEMS) is a rare disorder of unclear
pathogenesis which is probably mediated by i production of lambda light
chains from abnormal plasma cells.
Clinical presentation
• Progressive polyneuropathy.
• Organomegaly, especially hepatosplenomegaly and lymphadenopathy.
• Common endocrinopathies (84%) (multiple in 65%) include
hypogonadotrophic hypogonadism (70%—2/3 central, 1/3 p),
hypothyroidism (60%), hypoadrenalism (60%), diabetes mellitus (50%),
hyperprolactinaemia (20%).
• Monoclonal gammopathy.
• Skin changes.
Treatments
Include chemotherapy, irradiation, and surgery.
Further reading
Dispenzieri A, Gertz MA (2004). Treatment of POEMS syndrome. Curr Treat Options Oncol 5,
249–57.
586 CHAPTER 9 Inherited endocrine syndromes
MEN type 1
Definitions
Characterized by:
• Parathyroid tumours/hyperplasia.
• Anterior pituitary adenomas.
• Pancreatic neuroendocrine tumours.
A clinical diagnosis of MEN-1 is made by the presence of two out of three
of these tumours or one of these tumours in the context of a family his-
tory of MEN-1.
The prevalence of MEN-1 has been estimated at 1 in 10,000 of the
population.
Genetics
• MEN-1 is an autosomal dominant condition, with an estimated
penetrance of 99% by the age of 50 years.
• The MEN-1 gene is located on the long arm of chromosome 11
(11q13) and is a tumour suppressor gene. The function of the encoded
protein MENIN is under investigation.
• The mutations causing MEN-1 are inactivating in nature.
• More than 1,300 mutations in the MEN-1 gene have been described
in affected families, and there is no clear genotype–phenotype
correlation.
• Mutational analysis is available in the UK, although it is important to
note that up to 10% of patients will not have mutations identifiable
within the coding region of the MEN-1 gene.
Clinical features
• The disease has been reported to manifest itself from the ages of 5 to
80 years.
• MEN-1 is associated with a high mortality due to aggressive tumours
or biochemical sequelae of the tumours.
• p hyperparathyroidism is the commonest presenting feature in MEN-1
and occurs in 85–95% of patients with MEN-1. The next commonest
presenting features are insulinomas or prolactinomas.
• Pituitary adenomas are found in approximately 30% of MEN-1 patients
(see Table 9.3).
• The incidence of pancreatic endocrine tumours varies between 30%
and 80% in different series (see Table 9.4).
• Other lesions are also associated with MEN-1 (see Table 9.5).
MEN TYPE 1 587
Further reading
Brandi ML, Gagel AF, Angeli A, et al. (2001). Guidelines for diagnosis and therapy of MEN type 1
and 2. J Clin Endocrinol Metab 86, 5658–71.
Thakker RV, Newey PJ, Walls GV, et al. (2012). Clinical practice guidelines for multiple endocrine
neoplasia type 1 (MEN1). J Clin Endocrinol Metab 97, 2990–3011.
MANAGEMENT OF MEN TYPE 1 591
592 CHAPTER 9 Inherited endocrine syndromes
MEN type 2
Definitions
MEN-2 may be divided into three forms:
• MEN-2A comprises of familial medullary thyroid carcinoma (FMTC)
in combination with phaeochromocytoma and parathyroid tumours.
MEN-2A accounts for nearly 90–95% of all MEN-2 cases.
• MEN-2B is defined as the occurrence of FMTC in association with
phaeochromocytoma, mucosal neuromas, and a marfanoid habitus.
• FMTC may also occur in isolation.
• See Table 9.7 for classification.
Genetics
• MEN-2 is an autosomal dominant condition.
• The C-RET proto-oncogene, activating mutations of which cause
MEN-2, is located on the long arm of chromosome 10 (10q11.2).
• This gene encodes RET which is a transmembrane receptor with an
extracellular cysteine-rich domain and an intracellular tyrosine kinase
domain. The protein plays a role in the development of the neural
crest and the enteric nervous system.
• The C-RET proto-oncogene is also involved, via inactivating mutations,
in the aetiology of Hirschsprung’s disease.
• Different germline mutations cause different clinical syndromes. In
contrast to MEN-1, MEN-2 shows a strong genotype–phenotype
correlation.
• Approximately 50% of MEN-2A is caused by a codon 634 mutation,
and nearly all MEN-2B is caused by the Met918Thr mutation.
• Mutational analysis is available in the UK.
Clinical features—MEN-2A
MTC
• MTC is often the initial manifestation and is generally multifocal.
• Phaeochromocytoma and parathyroid disease typically develop later.
• Diagnosis of MTC requires histological analysis which reveals C cell
hyperplasia and stromal amyloid.
• Circulating calcitonin levels are generally elevated, but hypocalcaemia
is not seen.
• CEA is also elevated and is a useful tumour marker.
• With metastatic disease, diarrhoea is common (30%).
• Rarely, these tumours secrete ACTH, resulting in Cushing’s syndrome
due to ectopic ACTH secretion.
Phaeochromocytoma
• Usually presents later than MTC.
• Most commonly associated with codon 634 mutations.
• 50% will be bilateral, but malignancy is rare (<10%).
• These lesions must be excluded prior to surgery for any other
indication.
MEN TYPE 2 593
Primary hyperparathyroidism
• Generally results from hyperplasia of the glands.
• Mainly associated with codon 634 mutations, less commonly with
other RET mutations.
• Can present as early as 5 years with codon 634 mutations, and
commencement of early screening is recommended.
Clinical features—MEN-2B
• Mucosal neuromas can be found on the distal tongue, conjunctiva, and
throughout the gastrointestinal tract (may cause malabsorption).
• A marfanoid habitus is typically evident.
• MTC tends to present earlier than in MEN-2A and frequently follows a
more aggressive course.
594 CHAPTER 9 Inherited endocrine syndromes
Mutational analysis
• First- and second-degree relatives of patients with MEN-2 and genetic
analysis should be performed as early as possible in at-risk children so
that appropriate treatment can be planned early.
• Consider screening in patients with sporadic MTC or sporadic
phaeochromocytoma or in patients with mucosal neuromas or other
phenotypic features compatible with MEN-2B.
• Somatic RET oncogene mutations confer a worse prognosis in sporadic
medullary carcinoma.
Screening for MEN type 2
Biochemical and radiological screening is relevant for affected patients,
asymptomatic mutation carriers, and first- and second-degree relatives in
families with a clinical diagnosis of MEN-2, but where a mutation has not
been identified (see Table 9.8).
Further reading
Brandi ML, Gagel AF, Angeli A, et al. (2001). Guidelines for diagnosis and therapy of MEN type 1
and 2. J Clin Endocrinol Metab 86, 5658–71.
Carling T (2005). Multiple endocrine neoplasia syndrome: genetic basis for clinical management. J
Curr Opin Oncol 17, 7–12.
Constante G, Meringolo D, Durante C, et al. (2007). Predictive value of serum calcitonin levels for
preoperative diagnosis of medullary thyroid carcinoma in a cohort of 5817 consecutive patients
with thyroid nodules. J Clin Endocrinol Metab 92, 450–5.
Marx SJ (2005). Molecular genetics of multiple endocrine neoplasia types 1 and 2. J Nat Rev Cancer
5, 367–75.
Waguespack SG, Rich TA, Perrier ND, et al. (2011). Management of medullary thyroid carcinoma
and MEN2 syndromes in childhood. Nat Rev Endocrinol 7, 596–607.
Wirth LJ, et al. (2013). Case records of the Massachusetts General Hospital. Case 5-2013.
A 52-year-old woman with a mass in the thyroid. N Engl J Med 368, 664–73.
596 CHAPTER 9 Inherited endocrine syndromes
Table 9.9 Acid load test for diagnosis of renal tubular acidosis
Side effects Nausea
Procedure Normal breakfast
Fluid intake 200–300ml/h
08.00 empty bladder
Hourly collection of urine for pH over 10h
10.00 ammonium chloride capsules 0.1g/kg taken over 1h to
avoid gastric irritation
Sampling Urine hourly–pH
Electrolytes 10:00 14:00 18:00
Interpretation Urine pH should fall <5.2
Plasma bicarbonate should fall (ammonium chloride
absorbed)
Causes of hypercalciuria
With hypercalcaemia
• Primary hyperparathyroidism.
• Granulomatous disease.
• Vitamin D excess.
• Malignancy (rare as a cause of stone).
• Hyperthyroidism.
With normocalcaemia
• Distal renal tubular acidosis.
• Granulomatous disease.
• Cushing’s disease.
• Idiopathic hypercalciuria.
• Dent’s disease.
• Bartter syndrome.
• Hypocalcaemic hypercalciuria.
• Hereditary hypophosphataemic rickets.
• Acromegaly.
Further reading
Langman CB (2004). The molecular basis of kidney stones. Curr Opin Paediatr 16, 188–93.
Sakhaee K, et al. (2012). Clinical review. Kidney stones 2012: pathogenesis, diagnosis, and manage-
ment. J Clin Endocrinol Metab 97,1847–60.
Thakker RV (2004). Diseases associated with the extracellular calcium-sensing receptor. Cell
Calcium 35, 275–82.
Worcester EM, Coe FL. (2010). Clinical practice. Calcium kidney stones. N Engl J Med 363, 954–63.
Chapter 10 601
Endocrine surgery
Transsphenoidal surgery/craniotomy
Preoperative assessment
Confirm the following:
• Anterior and posterior pituitary function normal or on adequate
replacement:
• Short Synacthen® test (note both the 0 and 30min values), and/
or normal ACTH/9 a.m. cortisol. NB Patients with recent loss of
ACTH will have a normal response to SST, as adrenal atrophy will
not have evolved. If in any doubt, replace with glucocorticoids.
• FT4.
• LH, FSH, oestradiol, or testosterone.
• Prolactin.
• Serum and urine osmolality, and electrolytes if polyuric.
• Recent (<3 months) MRI pituitary.
• Formal visual field perimetry and visual acuity assessment.
• Document extraocular muscle movements.
• Urea and electrolytes (<1 week presurgery).
• Group and save serum.
• MRSA screen should be done at least 2 weeks prior to admission for
surgery if the patient has been in hospital within the last year.
• Record therapeutic options discussed with patient, including:
• Risks of surgery (e.g. CSF leakage, meningitis, bleeding, partial or
total hypopituitarism, including diabetes insipidus, and potential
effects on fertility and visual deterioration).
• Risk of recurrence requiring further surgery or radiotherapy.
• Warn patients that a sample of fat may be taken from their thigh or
abdomen for packing of the pituitary fossa.
• Reiterate the need for lifelong follow-up.
• Ensure antibiotic prophylaxis is given prior to surgery (the precise
regimen may vary, depending on the centre and patient, e.g.
flucloxacillin 500mg qds and amoxicillin 500mg tds PO/IV; use
erythromycin if penicillin-allergic). Some surgeons advocate a
prolonged course of antibiotics if CSF leakage occurs whilst others
recommend close surveillance and prompt treatment if concern
regarding possible meningitis (NB lower threshold for patients with
Cushing’s disease).
• MRSA +ve patients require prophylaxis with IV vancomycin.
• If steroid-deficient, steroid reserve is unknown, or if a patient has
Cushing’s disease (inadequate stress response), give hydrocortisone
20mg orally with morning premedication.
• If on levothyroxine preoperatively (for s hypothyroidism), change to
equivalent dose of T3.
TRANSSPHENOIDAL SURGERY/CRANIOTOMY 603
Post-operative
• Start hydrocortisone (20mg at 8 a.m./10mg at 12 noon/10mg at 6p.m.)
after surgery. Reduce to hydrocortisone 10mg/5mg/5mg on day 3
post-operatively.
• Watch for: headache, fever/photophobia/other signs of meningitis,
CSF leak, deterioration of vision, diplopia, bleeding, symptoms of
hyponatraemia.
• Fluid balance—watch for diabetes insipidus:
• Review the clinical status of the patient at regular intervals (?
euvolaemic/hypovolaemic/hypervolaemic).
• Record fluid input/output assiduously (NB fluid replacement in
theatre may be excessive; therefore, always include perioperative
fluids in fluid balance charts, and note sodium content).
• Check U&Es, plasma and urine osmolalities on a regular basis (at
least once daily and more frequently if clinical concerns).
• Post-operatively, restrict to 2L total fluid input (IV and PO).
• If patient becomes polyuric, i.e. >200mL/h for ≥3 consecutive hours
(in the context of a 2L/24h fluid restriction), then urgently check
plasma U&Es, plasma and urine osmolalities, and urinary sodium.
While waiting for results, allow free fluids; aim to replace the fluid
deficit.
• If diabetes insipidus confirmed by the results, give a single dose of
desamino-D-arginine vasopressin (desmopressin) (1 microgram SC).
• When fluid deficit has been replaced (usually orally), restart 2–3L
fluid restriction (again, include IV and PO routes).
• If polyuria recurs, treat as before.
• Regular U&Es, plasma and urine osmolalities required.
• If polyuria continues to recur up to and after 96h post-operatively,
consider regular DDAVP orally or intranasally, if possible (not if a
transsphenoidal approach is used).
• Hyponatraemia occurring 1 week after a transsphenoidal adenectomy
is most commonly due to SIADH; rarely, cerebral salt wasting may
occur. Check urinary sodium, and assess fluid status (cerebral salt
wasting causes very high urinary sodium and is associated with
dehydration and a high urine volume).
• Check for CSF leakage: bedside-test nasal fluid with Glucostix® and
send a sample for beta-transferrin/tau-protein (abundant in neurons)
for confirmation (+ve if CSF).
• If patients sent home on day 3 post-operatively, they should be advised
to watch for a fever, headache, excessive sensitivity to light, confusion,
drowsiness, vomiting, nasal discharge, deterioration of vision, excessive
urination (>200mL/h for ≥3 consecutive hours).
• In patients with Cushing’s disease, check 9 a.m. cortisol concentrations
on day 7, having omitted hydrocortisone dose on the evening before
and on the morning of the test. 9 a.m. cortisol <50nmol/L suggests cure;
occasionally, cortisol takes a few days to fall to undetectable levels.
• For all patients on day 7, recheck visual acuity and formal visual field
perimetry and eye movements, along with FBC, ESR, LFTs, U&Es,
604 CHAPTER 10 Endocrine surgery
plasma and urine osmolalities, glucose, TFTs (for those patients not
on T3).
• Comment on results:
• If FT4 and FT3 low, offer T3 and recheck TFTs in 6 weeks.
• If serum cortisol >450nmol/L, stop hydrocortisone.
• If serum cortisol 150–450nmol/L, stop hydrocortisone and cover
with steroids in times of stress.
• If serum cortisol <150nmol/L, continue on hydrocortisone.
• Recheck HPA axis 6 weeks post-operatively.
• Give information to patient, including advice on driving:
• For an ordinary driving licence (group 1, car or motorcycle), a
minimum horizontal field of 120 degrees is required as well as no
significant defect within the central 20 degrees. For a licence to
drive a group 2 vehicle (bus or lorry), then normal binocular field is
required. Taxi driver licence regulations vary locally.
• If an individual has normal visual fields and acuity and an
uncomplicated transsphenoidal operation, they may restart to drive
once recovered from the surgery (group 1 and group 2 licences).
• Following a craniotomy, group 2 licence holders are suspended
for 6 months; however, group 1 licence holders can restart driving
once recovered, providing their vision is satisfactory and there is no
history of seizures.
• All patients should inform their insurance company about the
pituitary tumour and pituitary surgery.
• Give information to patient on contact details for the Pituitary
Foundation support group and DVLA:
• The Pituitary Foundation, 17/18 The Courtyard, Woodlands,
Bradley Stoke, Bristol BS12 4NQ. Tel: 01454 201612.
• Medical Adviser, The Drivers’ Medical Branch, 2 Sandringham Park,
Swansea Vale, Llansamlet, Swansea SA6 8QD. Tel: 0870 0600 0301.
TRANSSPHENOIDAL SURGERY/CRANIOTOMY 605
606 CHAPTER 10 Endocrine surgery
Thyroidectomy
Preoperative
• Ensure euthyroidism. Surgery in the presence of poorly controlled
hyperthyroidism is associated with an increased post-operative
mortality (often by precipitation of a thyroid storm).
• If surgery is required in the presence of hyperthyroidism, give
potassium iodide (60mg 3× a day for 10 days); this reduces thyroid
hormone release and probably decreases perioperative blood loss.
The radiographic contrast agent iopanoic acid, which is rich in iodine,
provides a useful alternative and has the additional benefit of potently
inhibiting the 5-deiodinase, thus reducing T4 to T3 conversion. Oral
propranolol (40–120mg 3× a day) reduces clinical manifestations of
thyrotoxicosis. Steroids (e.g. hydrocortisone 100mg IV 4× a day) can
be used to decrease T4 to T3 conversion.
• Check vocal cord function by indirect laryngoscopy.
• Warn of post-operative risks: recurrent laryngeal nerve damage <1%,
keloid scarring, haemorrhage, permanent hypoparathyroidism <0.5%,
and hypothyroidism (10% of partial thyroidectomy patients).
Post-operative
• Risk of haemorrhage in first 24h, particularly major haemorrhage deep
to the strap muscles leading to airway compression. Watch for stridor,
respiratory difficulties, and wound swelling. Drainage from wound
drains is unhelpful. Treat by evacuating the haematoma; consider
intubation or a tracheostomy. Clip removers and artery forceps should
be kept to hand on the ward.
• Recurrent laryngeal nerve damage is permanent in <1% and
transient in 2–4%. Patient’s voice is often husky for about 3 weeks
post-operatively and may be treated with lozenges and humidified air.
• Symptomatic unilateral damage can be treated by stabilization of the
affected cord in adduction by submucosal Teflon® injection under
direct laryngoscopy.
• Bilateral damage leads to unopposed adductor action of the
cricothyroid muscle which causes glottis closure and airway
obstruction. Treatment involves reintubation, paralysis, hydrocortisone
(100mg 4× a day IM for oedema), and extubation at 24h—if that fails, a
tracheostomy should be performed.
• If recurrent laryngeal nerve damage is persistent at 9 months, an
attempt can be made to resuture the nerve.
• Monitor calcium. Transient hypoparathyroidism is usually evident
within 7 days (for treatment, b see Hypoparathyroidism, p. 478).
• Patients undergoing thyroidectomy are at high risk of post-operative
nausea and vomiting.
• Following total thyroidectomy for malignancy, the patient should be
converted to T3, which should be stopped at least 10 days prior to
the post-operative radioiodine uptake scan to allow the TSH to rise
(b see Follow-up of papillary and FTC, p. 100). Alternatively,
recombinant TSH can be used for the scan.
THYROIDECTOMY 607
Parathyroidectomy
Parathyroidectomy of one or two glands undertaken for p hyperpar-
athyroidism may result in transient and self-limiting hypocalcaemia. Total
parathyroidectomy (e.g. for MEN-1 or as part of surgical management of
advanced head and neck malignancy) may be complicated by severe and
permanent hypocalcaemia which may be very difficult to manage.
Post-operative care for patients undergoing
parathyroidectomy of 1–2 glands
• Check calcium, phosphate, magnesium, albumin on the evening of
surgery and daily thereafter. Calcium begins to fall post-operatively
after about 4–12h; the nadir is usually reached by 24h. Calcium may
recover spontaneously; however, one-third of patients will require
calcium support perioperatively. With the advent of minimally invasive
parathyroidectomy and short hospital stays (<24h), many centres
advocate prophylactic calcium and vitamin D replacement in all cases
in the immediate aftermath of surgery, which is continued until the
patient is reviewed 1–2 weeks later in the outpatient clinic.
• Symptoms of hypocalcaemia (mainly due to neuromuscular
irritability): perioral paraesthesiae, Chvostek’s sign, Trousseau’s sign,
tetany, laryngospasm, bronchospasm, seizures, prolonged QT interval
on ECG, extrapyramidal movement disorders, and delirium. Calcium
levels often <1.75mmol/L before symptoms manifest, although rapid
changes in calcium result in more pronounced symptomatology.
• Magnesium deficiency is common due to previous hyperparathyroidism
(causes renal wasting of magnesium). Chronic magnesium deficiency
impairs release of PTH and causes functional hypoparathyroidism and
hypocalcaemia.
Causes of hypocalcaemia post-1–2 gland removal
• ‘Functional hypoparathyroidism’ common. Causes: delayed recovery
of the other parathyroid glands due to long-term suppression;
parathyroid gland ischaemia; parathyroid gland ‘stunning’ by
intraoperative handling; hypomagnesaemia. PTH level will be
detectable; phosphate should be normal. Usually spontaneously
improves over days to weeks. Management of symptomatic
hypocalcaemia (Ca usually <1.8mmol/L) with calcium (up to 2g/day in
divided doses). Add in vitamin D/vitamin D metabolites if persistent
hypocalcaemia. Replace magnesium, as necessary. Gradual withdrawal
of therapy to assess recovery.
• ‘Hungry bone syndrome’ due to extensive skeletal remineralization
once skeleton released from PTH excess. Ongoing i ALP, d calcium,
d PO4, d Mg. PTH levels may be normal or high. Pre-existing vitamin
D deficiency will exacerbate hypocalcaemia. May require large
doses of calcium and vitamin D/vitamin D metabolites for weeks to
months.
• Permanent hypoparathyroidism. Rare (<2% of cases). Check PTH level
after day 3; level will be undetectable (<1pg/mL). Replace with oral
calcium and vitamin D/vitamin D metabolites long-term.
PARATHYROIDECTOMY 609
• Other complications:
• Recurrent laryngeal nerve palsy (<1%).
• Failure to correct hypercalcaemia.
• Overall, both minimally invasive and conventional parathyroidectomy
are very safe operations, with low morbidity and mortality.
Calcium management following total
parathyroidectomy
• Hypocalcaemia is inevitable unless management instituted.
• Pre-emptive treatment is worth considering (e.g. 1A-calcidol 1–2
micrograms/day—this dose may be insufficient to completely prevent
hypocalcaemia but may prevent life-threatening hypocalcaemia and is
unlikely to cause serious toxicity in the short term).
• Acute management in patients with life-threatening hypocalcaemia (e.g.
Trousseau’s sign, laryngospasm, seizures):
• 10mL of 10% calcium gluconate, diluted 1 in 10 in normal saline
or glucose 5%, infused into large vein over 10min. Monitor cardiac
rhythm.
• Repeat, as necessary, to control acute emergency.
• Patients will need ongoing calcium replacement: 100mL of 10%
calcium gluconate in 1L of 5% glucose or 0.9% sodium chloride,
infused over 24h (monitor calcium regularly (4–6-hourly), and
adjust rate as necessary).
• Start oral vitamin D analogues and oral calcium.
Vitamin D analogues (1A-calcidol; calcitriol)
• Potent and effective in acute hypocalcaemia due to rapid correction
of calcium. Short half-lives allows for rapid and careful titration of
dose in response to calcium levels. Narrow therapeutic window
(but hypercalcaemia much shorter lived if it develops).
• Dose for dose, calcitriol twice as efficacious as 1A-calcidol (i.e.
1 microgram calcitriol equivalent to 2 micrograms 1A-calcidol).
1A-calcidol can be given down an NG tube.
• Starting doses usually high (e.g. 4–8 micrograms/day of 1A-calcidol in
divided doses), rapidly weaned to maintenance doses (typically 1–2
micrograms/day 1A-calcidol).
• Reassess often (at least every 1–2 days) in early stages of management.
Longer-term options include ergocalciferol or colecalciferol, but
hypercalcaemia will be more prolonged if it develops.
Calcium
• 1–2g/daily in divided doses. A maximum daily absorbable dose of
calcium is probably 3g day.
• Absorption may vary from different types of calcium salts and may be
greater when calcium is given away from food.
• The long-term aim is to manage without calcium, just on vitamin
D/vitamin D metabolites.
Long-term goal of management is to prevent symptoms of hypocalcaemia
without toxicity. Aim for lower half of normal range (2.0–2.3mmol/L), with
normal urinary calcium excretion (to minimize risk of nephrolithiasis and
nephrocalcinosis).
610 CHAPTER 10 Endocrine surgery
Phaeochromocytoma
Preoperative
• Check for bilateral disease or metastases: MRI chest and abdomen, and
an MIBG scan (10% multiple).
• Ensure adequate A- and B-blockade once the diagnosis is made.
• Start B-blockade only AFTER patient has been adequately A-blocked
(unopposed B-blockade can lead to marked vasoconstriction,
ischaemic damage, and hypertension).
• A-blockade. Start phenoxybenzamine (10–20mg PO, 3–4x day)—an
irreversible A-blocker. Adequacy of dose assessed by monitoring
haematocrit and postural BP drop (reflex vasodilation). Side effects
include dizziness, lethargy, and ankle swelling. Some centres use
doxazosin; others are concerned that this does not offer both A1-
and A2-blockade.
• Start treatment at least 1 week before surgery, ideally >3 weeks.
• B-blockade. Start propranolol (20–80mg PO, 3x day) when patient
adequately A-blocked. Titrate to maximum tolerated or a total
dose of 240mg/day in divided doses.
• Control BP: A- and B-blockers often sufficient; if not, add a calcium
channel blocker or an ACE inhibitor; A-methyltyrosine 1–4g/day (a
false catecholamine precursor which inhibits tyrosine hydroxylase,
the rate-limiting step for catecholamine synthesis) is rarely used to
control BP.
• Some centres advocate the use of additional IV phenoxybenzamine
(0.5–1.0mg/kg in 250mL 5% dextrose, given over 2h) for 3 days prior to
surgery (titrate the dose according to BP; often, 0.5mg/kg is sufficient).
• Monitor haemoglobin/haematocrit (because of haemodilution,
preoperative blood transfusions may be necessary) and postural BP.
• Group and save serum, and cross-match 2 units of blood.
• Ensure that the patient is well hydrated (if necessary, use an IV infusion
of saline) prior to going to theatre.
Post-operative
• Stop A- and B-blockers.
• Watch for d BP: sudden withdrawal of catecholamines leads
to marked arterial and venous dilatation. This is worsened by
inadequate volume loading and should initially be treated with volume
replacement rather than by pressor agents.
• If hypertension persists 2 weeks post-operatively, then residual tumour
or metastases must be considered.
• Long-term monitoring required, as approximately 14% recur.
• If bilateral adrenalectomy performed, see next section.
PHAEOCHROMOCYTOMA 611
Bilateral adrenalectomy
• Give hydrocortisone (100mg, 4x day IM) post-operatively; this will
provide adequate mineralocorticoid as well as glucocorticoid cover.
Continue this until eating and drinking (often <48h). Monitor U&Es.
• From day 3, give hydrocortisone PO (double usual replacement dose,
e.g. 20mg/10mg/10mg), and add fludrocortisone PO (100 micrograms
daily).
• Long-term replacement with hydrocortisone 10mg/5mg/5mg, and
fludrocortisone 50–150 micrograms daily.
Phaeochromocytoma in pregnancy (b see p. 447)
• Rare condition; may present as paradoxical supine hypertension, with
pressure from the gravid uterus causing release of catecholamines, and
normal blood pressure in the supine and erect positions.
• Start A-blockade, then B-blockade; phenoxybenzamine can cross the
placenta but is generally safe for the fetus (may cause perinatal CNS
depression and transient hypotension). Propranolol has been reported
to be associated with intrauterine growth retardation, fetal bradycardia
and hypoglycaemia, and premature labour.
• Surgery is more controversial, although some authors advocate
surgery in the first and second trimesters up to 24 weeks’ gestation.
• Caesarian section is advocated, with a combined tumour resection.
Vaginal delivery carries a significant maternal risk.
Chapter 11 613
Endocrinology
and ageing
• Fat body mass increases more than lean body mass with age; thus,
there is i aromatization of androgens to oestrogens. The effects of
this are unclear.
• Hypoandrogenism may also result from hyperprolactinaemia due to
pituitary/hypothalamic disease, renal dysfunction, hypothyroidism,
drugs (psychotropic and anti-dopaminergic agents); all more common
in the elderly population.
Testosterone therapy (b p. 378)
• Few small studies in elderly ♂, either as replacement in patients with
clear hypogonadism or in healthy ♂.
• Data point towards a +ve effect on well-being, muscle mass and
strength, and d fat mass in elderly patients, with greatest effect in
patients with clear hypogonadism.
• Risk of s polycythaemia, liver dysfunction (particularly if testosterone
taken orally), prostatism, exacerbation of prostate adenocarcinoma,
and possibly dyslipidaemia.
Erectile dysfunction (b p. 388)
• Common in elderly ♂. 50% of ♂ >60 have erectile dysfunction; 90%
of these ♂ have concurrent medical problems or are on medication
potentially causing impotence.
• Aetiology often multifactorial:
• Atherosclerosis—commonest cause with both macro- and
microvascular disease.
• Penile denervation—autonomic neuropathy (most commonly due
to diabetes mellitus); pelvic surgery (including prostatectomy—30%
of ♂ >75 develop erectile dysfunction after prostatectomy
(compared with 7% of younger ♂ after prostatectomy)).
• Drugs (B-blockers, calcium channel antagonists, other
antihypertensive agents, psychotropic drugs).
• Psychogenic.
Delayed/absent ejaculation
• i common with age due to autonomic nerve dysfunction, drugs,
previous surgery, and usually the harbinger of erectile dysfunction.
• Evaluation similar to that of younger patients (b see Evaluation of
erectile dysfunction, p. 390).
• Management similar to younger patients, with caveat that
phosphodiesterase inhibitors may interact with nitrates and
antihypertensive agents.
Fertility
• Testicular morphology, semen production, and testicular
steroidogenesis are maintained into old age.
• There is some evidence for a small increase in specific genetic
disorders in the children of older men.
622 CHAPTER 11 Endocrinology and ageing
Endocrinology aspects
of other clinical or
physiological situations
Hypoglycaemia 628
Symptoms of hypoglycaemia 630
Investigations of hypoglycaemia 632
Management of hypoglycaemia 634
Postprandial reactive hypoglycaemia (PRH) 636
Mastocytosis 638
Cancer 640
Endocrine sequelae of survivors of childhood cancer 642
Syndromes of ectopic hormone production 646
SIADH due to ectopic vasopressin production 648
Humeral hypercalcaemia of malignancy 650
Cushing’s syndrome due to ectopic ACTH production 652
Endocrine dysfunction and HIV/AIDS 654
Liver disease and endocrinology 658
Renal disease and endocrinology 660
Endocrinology in the critically ill 662
Syndromes of hormone resistance 664
Differential diagnosis of possible manifestations of endocrine
disorders 666
Stress and the endocrine system 670
Endocrinology of exercise 672
Complementary and alternative therapy and endocrinology 674
Alternative therapy used in patients with diabetes mellitus 676
Alternative therapy used in menopause 678
Alternative therapy used by patients with osteoporosis 680
Miscellaneous alternative therapy 682
628 CHAPTER 12 Other clinical/physiological situations
Hypoglycaemia
Definition (Whipple’s triad)
• Plasma glucose of <2.2mmol/L, associated with
• Symptoms of neuroglycopenia, and
• Reversal of symptoms with correction of glucose levels.
Epidemiology
Uncommon in adults, apart from patients with diabetes being treated with
certain agents, either alone or in combination (e.g. insulin, sulfonylureas).
Pathophysiology
Physiology of glucose control
Plasma glucose concentrations are usually kept within narrow limits
(~3.3–5.6mmol/L), providing an uninterrupted supply of glucose to the
brain (which can consume up to 50% of hepatic glucose output).
The liver is the major regulator (80–85%) of circulating blood glucose
concentrations in healthy individuals and responds to changes in circulat-
ing insulin, GH, cortisol, glucagon, and adrenaline.
• Postprandial state: hepatic glucose production is inhibited by i plasma
glucose and i insulin concentrations.
• Fasting state: plasma glucose d with consequent d in insulin secretion,
stimulating hepatic glucose efflux (due to i cortisol, GH, and
glucagon).
Mechanisms of hypoglycaemia
• Excessive/inappropriate action of insulin (or IGF-1): inhibiting hepatic
glucose production, despite adequate glycogen stores, while peripheral
glucose uptake is enhanced.
• Impaired neuroendocrine response with inadequate counter-regulatory
response (e.g. cortisol) to insulin.
• Impairment of hepatic glucose production due to either structural
damage or abnormal liver enzymes.
Classification of hypoglycaemia
Traditionally, hypoglycaemia has been classed as fasting or postprandial.
• Fasting hypoglycaemia: occuring several hours (typically >5h) after food
(e.g. early morning, following prolonged fasting or exercise).
• Postprandial (reactive) hypoglycaemia: occuring 2–5h after food.
Causes of hypoglycaemia can fit into both categories, so another clas-
sification has been proposed, based on whether the patient is unwell (see
Box 12.1.)
HYPOGLYCAEMIA 629
Symptoms of hypoglycaemia
• See Table 12.1 for classification of symptoms.
• Adrenergic symptoms have been identified at arterialized plasma
glucose concentrations of 3.3mmol/L (equivalent to a venous plasma
concentration of 3.1mmol/L).
• Neuroglycopaenic symptoms have been identified at arterialized
plasma glucose concentrations of 2.8mmol/L (equivalent to a venous
plasma concentration of 2.6mmol/L).
• EEG changes occur at 2.0mmol/L.
• The majority of symptoms of acute hypoglycaemia are adrenergic,
but neuroglycopaenic symptoms occur with subacute and chronic
hypoglycaemia.
• The rate of decrease in the plasma glucose concentration does not
influence the occurrence of the symptoms.
• Patients with recurrent hypoglycaemia may not get symptoms until
glucose concentrations are very low (so-called ‘hypo unawareness’),
whilst patients with poorly controlled diabetes mellitus may
experience hypoglycaemic symptoms at ‘normal’ blood glucose levels.
Investigations of hypoglycaemia
• Glucose (BM) strip: unreliable for low glucose concentrations.
• Liver and renal function tests.
• Blood ethanol concentration.
• Synacthen® test.
• Insulin, C-peptide, pro-insulin, and glucose during hypoglycaemia
(see Table 12.2).
• Inappropriately elevated insulin in presence of hypoglycaemia suggests
insulinoma, self-administration of insulin/sulfonylurea, post-gastric
bypass hypoglycaemia, non-insulinoma pancreatogenous hypoglycaemia
syndrome, or insulin autoimmune hypoglycaemia.
• Absence of C-peptide, but the presence of insulin, during hypoglycaemia
suggests exogenous insulin administration or raised IGF-II.
• Insulinomas often associated with elevated pro-insulin:insulin ratio.
• Consider assay for presence of sulfonylureas.
• Fasting B-hydroxybutyrate (elevated in most causes of hypoglycaemia
but suppressed if insulin present, e.g. insulinoma, self-administration of
insulin, or sulfonylureas).
• Consider IGF-I and II and pro-IGF-II. IGF-II may be normal in non-islet
cell hypoglycaemia, but this is in association with suppressed IGF-I
and GH; usual IGF-II:IGF-I ratio 3:1, ratio >10 seen in non-islet cell
hypoglycaemia. Tumours tend to secrete pro-IGF-II, leading to a raised
pro-IGF-II:IGF-II ratio.
• Chest and abdominal radiographs/CT.
• Consider insulin and insulin receptor antibodies.
Further investigation of fasting hypoglycaemia
• 15h fast:
• Measure plasma glucose and insulin after fasting for 15h.
• Glucose <2.2mmol/L and insulin >5mU/L is inappropriate.
• Good screening test if repeated 3 times.
• 75% of patients with an insulinoma will develop hypoglycaemia
within 18h of beginning a fast.
• 72h fast:
• The most reliable test for hypoglycaemia (detects 98% patients with
insulinoma, compared with >70% at 24h).
• The patient should remain hydrated and active.
• Measure plasma glucose, insulin, C-peptide, and pro-insulin
6-hourly (unless the patient is symptomatic or the glucose level is
<3.5mmol/L when measurements are made every 1–2h); the test is
terminated if the laboratory glucose <2.2mmol/L or after 72h.
• B-hydroxybutyrate should be measured at the end of the fast (its
presence makes insulinoma unlikely).
• C-peptide suppression test:
• Rationale: insulin administration to induce hypoglycaemia should
suppress endogenous insulin secretion. C-peptide serves as a
measure of endogenous insulin secretion and is suppressed during
hypoglycaemia to a lesser degree in people with an insulinoma than
in normal persons.
INVESTIGATIONS OF HYPOGLYCAEMIA 633
Management of hypoglycaemia
Acute hypoglycaemia
• If conscious:
• 15–20g oral carbohydrate (ideally food and a sugary drink) should
be administered as soon as possible.
• Dextrogel®/Glucogel® (formerly known as Hypostop Gel®), a
glucose-containing gel which is absorbed by the buccal mucosa, may
be used in drowsy, but conscious, individuals.
• If unconscious:
• 75–80mL of 20% glucose intravenously into a large vein (over
10–15min), followed by a saline flush as the high concentration
of glucose is an irritant and may even lead to venous thrombosis.
A maintenance infusion of 5% or 10% dextrose is often required
thereafter, especially if there is an ongoing risk of recurrent
hypoglycaemia (e.g. overdose of a long-acting insulin/analogue).
• 1mg glucagon IM may be administered if there is no IV access.
This increases hepatic glucose efflux, but the effect only lasts for
30min, allowing other means of blood glucose elevation (e.g. oral)
before the blood glucose falls again. It is ineffective with hepatic
dysfunction and if there is glycogen depletion, e.g. ethanol-related
hypoglycaemia, and is relatively contraindicated in patients with
known insulinoma, as it may induce further insulin secretion.
Glucagon is ineffective if given within 3 days of a previous dose of
glucagon.
• s cerebral oedema may complicate hypoglycaemia and should be
considered in cases of prolonged coma despite normalization of
plasma glucose. Mannitol and/or dexamethasone may be helpful.
Recurrent chronic hypoglycaemia
If definitive treatment of the underlying condition is unsuccessful or
impossible, symptoms may be alleviated by frequent (e.g. 4-hourly) small
meals, including overnight. Diazoxide, administered by mouth, is useful
in the management of patients with chronic hypoglycaemia from excess
endogenous insulin secretion due to an insulinoma or islet cell hyperplasia.
MANAGEMENT OF HYPOGLYCAEMIA 635
636 CHAPTER 12 Other clinical/physiological situations
Management
Diet
• Frequent, small, low-carbohydrate, high-protein meals.
• Avoid rapidly absorbed carbohydrates.
• Avoid sugary drinks, especially in combination with alcohol.
• Addition of soluble dietary fibres, e.g. 5–10g guar gum or pectin or
hemicellulose per meal, delays absorption and lowers the glycaemic
and insulinaemic indices (especially effective in rapid gut transit time).
Drugs
• Acarbose, an intestinal A-glucosidase inhibitor, delays sugar and starch
absorption, thus reducing the insulin response to a meal.
• Metformin can be useful, 500mg with meals.
• Supplemental chromium is reported to downregulate B-cell activity
and increase glucagon secretion.
• In exceptional cases, with debilitating PRH, diazoxide (side effects—
water retention, hypertrichosis, digestive disorders) or somatostatin
analogues may be required.
• Propranolol and calcium antagonists have been used; however,
controlled studies are lacking.
Further reading
Brun JF, Fedou C, Mercier J (2000). Postprandial reactive hypoglycaemia. Diabet Metab 26, 337–51.
Cryer PE (1997). Hypoglycemia: pathophysiology, diagnosis, and treatment. Oxford University
Press, New York.
Cryer PE, Axelrod L, Grossman AB, Heller SR, Montori V, Seaquist ER, Service FJ (2009). Evaluation
and management of adult hypoglycaemic disorders: an Endocrine Society clinical practice guide-
line. J Clin Endocrinol Metab 94, 709–28.
de Groot, et al. (2009). Non-islet cell tumour-induced hypoglycaemia: a review of the literature
including two new cases. Endocr Relat Cancer 14, 979–93.
Gama R, Teale JD, Marks V (2003). Clinical and laboratory investigation of adult spontaneous
hypoglycaemia. J Clin Path 56, 641–6.
Service FJ (1995). Hypoglycaemic disorders. N Engl J Med 350, 2272–9.
638 CHAPTER 12 Other clinical/physiological situations
Mastocytosis
Background and definitions
Mastocytosis is a heterogenous group of disorders characterized by the
pathological accumulation of mast cells in tissues. The main types of mas-
tocytoses are cutaneous and systemic. Systemic mastocytosis is frequently
diagnosed but seldom established as a differential diagnosis in the work-up
of flushing (sensation of warmth, usually on the face and neck, due to vaso-
dilation and increased cutaneous circulation). Flushing due to neurogenic
stimuli are likely to be associated with sweating (wet flushing), and flush-
ing due to vasodilator substances associated with ‘dry’ flushing. Systemic
mastocytosis usually features in the latter category.
See Box 12.2 for causes of raised tryptase.
Classification
Mastocytosis is broadly classified into cutaneous and systemic mastocyto-
sis (see Box 12.3).
Clinical features
• Urticaria pigmentosa and dermographism are the usual cutaneous
manifestations (exacerbated when rubbed—Darier’s sign).
• Systemic manifestations include flushing, anaphylaxis, abdominal pain,
and diarrhoea.
• Anaemia, hepatosplenomegaly, peptic ulceration, and steatorrhoea can
occur due to mast cell infiltration.
Epidemiology
• It is a rare disorder.
• Affects both sexes equally.
• Children usually have cutaneous mastocytosis, and adults the systemic
variety.
Investigations
• Serum tryptase, which is predominantly produced in mast cells, is
usually elevated.
• Skin biopsy stained with Giemsa and immunohistochemical staining for
tryptase and c-kit (mast cell growth factor receptor).
• Bone marrow biopsy is warranted for adults with evidence of
mastocytosis to diagnose systemic mastocytosis.
MASTOCYTOSIS 639
Treatment
• Avoidance of triggers (e.g. alcohol, aspirin).
• Treat anaphylaxis as a medical emergency.
• Preparation of patients appropriately before surgery and other medical
procedures with antihistamines, glucocorticoids, H2 blockade, and
montelukast.
• Antihistamines, such as cetirizine, fexofenadine, can be used to reduce
flushing and itching.
• H2 blockers and proton pump inhibitors, such as ranitidine and
omeprazole, can help with abdominal symptoms, such as pain and
heartburn.
• In patients who are not intolerant of NSAIDs, aspirin may help reduce
flushing.
• Additional antileukotriene agents, such as montelukast, may help some
with ongoing symptoms.
• Other treatment options, including tyrosine kinase inhibitors, have
been assessed, but their effectiveness is not convincing.
Further reading
Liu AY, et al. (2010). Clinical problem-solving. A rash hypothesis. N Engl J Med 363, 72–8.
Murali MR, et al. (2011). Case records of the Massachusetts General Hospital. Case 9-2011.
A 37-year-old man with flushing and hypotension. N Engl J Med 364, 1155–65.
640 CHAPTER 12 Other clinical/physiological situations
Cancer
Chemotherapy and radiotherapy may have endocrine effects.
Anticancer chemotherapy
There are three types of anticancer chemotherapeutic agents:
• Cytotoxics. These have no direct hormonal sequelae. Alkylating agents
are more likely to induce permanent ♂ sterility (without affecting
potency), and, in ♀, they may induce premature menopause which
may increase the likelihood of osteoporosis.
• Immunomodulators. Prednisolone in excess causes Cushing’s
syndrome, and acute withdrawal may precipitate adrenal insufficiency.
Cyclophosphamide, in particular, may cause early menopause. Thyroid
dysfunction has been reported rarely with tacrolimus and interferon
therapy.
• Hormones:
• Progestagens are used in breast cancer; of these, megestrol acetate
has potent glucocorticoid activity and thus may cause Cushing’s
syndrome in excess or adrenal insufficiency if abruptly withdrawn.
• Aromatase inhibitors, such as aminoglutethimide, may cause adrenal
insufficiency, and corticosteroid replacement is necessary.
• Trilostane, which inhibits 3B-hydroxysteroid dehydrogenase, may
also cause adrenal insufficiency.
• Gonadorelin analogues, used for prostatic cancer and breast cancer,
cause an initial increase in LH levels and then suppression and cause
side effects similar to orchidectomy in ♂ and the menopause in ♀.
• Antiandrogens, used in prostatic cancer, have predictable side
effects, such as gynaecomastia, hot flushes, impotence, and impaired
libido.
Radiotherapy
• Cranial radiotherapy, whose field encompasses the hypothalamo–
pituitary area, may result in hypopituitarism (b see Chapter 2,
Hypopituitarism, p. 202). The most radiosensitive axis is the GH axis,
followed by the FSH/LH axis, then the ACTH and TSH axes. Even low
radiation doses of <40Gy can affect the GH axis. Panhypopituitarism is
generally seen with doses of >60Gy, doses often used in the presence
of nasopharyngeal or skull base tumours.
• Head and neck irradiation may result in hypothyroidism and
hypoparathyroidism.
• After 5 or more years of follow-up, 50% of patients treated with
radiotherapy only for laryngeal and pharyngeal carcinoma will
develop hypothyroidism; combined surgery and radiotherapy results
in roughly 90% of patients developing hypothyroidism. The rates for
hypoparathyroidism were 88% and 90%, respectively.
• Radiotherapy affects the testes dose-dependently. Fertility is affected
much more than androgen-synthesizing capacity so that most ♂ have
normal testosterone levels unless given testicular doses >20–30Gy.
• The effects of radiotherapy to both ovaries are amplified with age.
Premature menopause may be elicited by doses >10Gy.
CANCER 641
642 CHAPTER 12 Other clinical/physiological situations
Clinical assessment
• Growth velocity.
• Weight and BMI.
• Sitting height (radiotherapy can cause vertebral dysplasia).
• Arm span.
• Pubertal assessment for evidence of precocious puberty (precocious
puberty can result in a falsely reassuring growth spurt).
Investigations
The diagnosis can be difficult to make. GHD is suspected when a
decreased growth velocity is found over a 6-month period. The diagnosis
is made using a combination of clinical features and investigations.
• Bone age to assess skeletal maturation.
• Insulin tolerance test (b see p. 114).
• GHRH with arginine may lack sensitivity in patients who have received
radiotherapy.
• IGF-1 and IGFBP-3 are not reliable markers of GH secretion following
radiotherapy or those with CNS lesions.
Treatment
• There is no clear evidence of either increased risk of recurrence of
the original tumour or death associated with GH therapy, despite the
anti-apoptotic and pro-proliferative effects of GH and IGF-1.
• There is evidence to suggest an increased risk of a secondary tumour
in survivors of acute leukaemia. Meningiomas are the commonest
second neoplasms.
• GH therapy in survivors of childhood cancers for adult GHD has
been shown to improve quality of life and modest improvements in
metabolic parameters.
• See section on growth hormone replacement (b see p. 134).
Thyroid abnormalities
Thyroid abnormalities are common endocrine sequelae in survivors of
childhood cancers.
Primary hypothyroidism
Primary hypothyroidism can develop up to 25 years after radiotherapy.
It is usually seen after mantle irradiation for Hodgkin’s lymphoma and
craniospinal irradiation for HS tumours. It is more likely to occur in girls.
Long-term surveillance with thyroid function tests, early detection, and
treatment are important.
Central hypothyroidism
Rare and usually associated with irradiation to the hypothalamic/pituitary
area of doses >30Gy. There is a small association between chemotherapy
and central hypothyroidism.
Hyperthyroidism
Much rarer than hypothyroidism and associated with doses of ≥35Gy to
the thyroid.
644 CHAPTER 12 Other clinical/physiological situations
Thyroid neoplasia
There is an increased likelihood of developing papillary and follicular can-
cers of the thyroid after radiotherapy. Children treated before the age of
10 and with doses between 20 and 30Gy are at highest risk. There can
be a very long latent period before the development of thyroid cancers;
therefore, ongoing long-term follow-up is essential.
Thyroid cancers secondary to radiotherapy do not behave more aggres-
sively than those occurring in people without radiotherapy.
Disruption of puberty
Puberty can be affected in survivors of childhood cancers in terms of pre-
cocious puberty, accelerated rate of progression through puberty, delayed
progression through puberty, or hypogonadotrophic hypogonadism.
Precocious puberty (see b p. 538)
Precocious puberty (<8 years for girls and <9 years for boys) is caused
by premature activation of the hypothalamo–pituitary–gonadal axis and
is associated with irradiation. It is more likely in girls and those who are
overweight and is associated with radiotherapy at an early age.
Clinical assessment
• Height and weight on growth charts, assessing growth velocity.
• An early sign of precocious puberty is an increase in growth velocity.
However, this can be masked either by concomitant GHD or
hypothyroidism.
• Tanner staging—in girls, evidence of breast development <8 years
is a sign of precocious puberty; in boys, however, testicular volume
is an unreliable marker, as there may be concomitant seminiferous
dysfunction.
• A bone age on X-ray of >2 SDs for the chronological age is a
consistent finding.
Management
Delaying the progression of puberty with GnRH agonists results in better
outcomes for height, especially in conjunction with GH where appropriate
for GHD (b see section on Precocious puberty, p. 538).
Hypogonadotrophic hypogonadism
Gonadotrophin deficiency is less common than GH deficiency and is
dependent on the dose of radiotherapy received to the hypothalamic–
sellar area. Whether chemotherapy alone results in gonadatrophin defi-
ciency is uncertain.
Assessment and management
b see section on delayed puberty, p. 542.
ENDOCRINE SEQUELAE OF SURVIVORS OF CHILDHOOD CANCER 645
Humeral hypercalcaemia
of malignancy
Hypercalcaemia is a common complication of malignancy; may be due
to ectopic hormone secretion (PTHrP; rarely 1,25(OH)2 colecalciferol);
cytokine and inflammatory mediators that activate osteoclastic bone
resorption (such as IL-6 and RANK-L production by myeloma cells); or
due to bone destruction by metastases.
Parathyroid hormone-related peptide (PTHrP)
• PTHrP binds to and activates PTH/PTHrP receptor type 1, resulting in
osteoclast-mediated bone resorption and reduced renal excretion of
calcium.
• The biochemical picture of hypercalcaemia and hypophosphataemia
may be indistinguishable from p hyperparathyroidism. However, PTH
levels are suppressed in PTHrP-mediated hypercalcaemia (NB p
hyperparathyroidism can coexist with malignancy).
• PTHrP secretion by metastatic cells within bone also causes
hypercalcaemia by causing local osteolysis.
• PTHrP can be measured directly and is elevated in 80% of cancer
patients with hypercalcaemia.
• Tumours that metastasize to bone are more prone to produce PTHrP
than tumours that do not metastasize to bone (50% of p breast cancer
express PTHrP, compared with 92% of metastases of breast cancer to
bone). This may be due to induction of PTHrP secretion by the bone
microenvironment; alternatively, PTHrP production by tumour cells
may enhance their ability to metastasize to bone.
• For tumours associated with humeral hypercalcaemia, b see
Table 12.3, p. 646.
Management
• As per normal management of hypercalcaemia (b see Other causes
of hypercalcaemia, p. 469).
• Glucocorticoids may be particularly effective in treatment of
hypercalcaemia associated with malignancy. This may be due to direct
effects of glucocorticoids on the tumour cells (e.g. haemopoietic
malignancies) and/or because of downregulation of production of
1,25(OH)2 colecalciferol.
• Bisphosphonates may also have antitumour effects in myeloma as well
as control osteoclastic destruction of bone.
HUMERAL HYPERCALCAEMIA OF MALIGNANCY 651
652 CHAPTER 12 Other clinical/physiological situations
Gonads
Males
• SHBG concentrations are often elevated; this can lead to normal total
testosterone concentrations but low free testosterone concentrations.
• Testosterone deficiency common in ♂ patients with AIDS (6% of
patients with asymptomatic HIV infection, compared with 50% of
patients with AIDS).
• Hypogonadism is associated with wasting, d muscle mass, fatigue, loss
of libido, and impotence. Hypogonadism may be p or s; up to 75%
of patients with hypogonadism have low or inappropriately normal
gonadotrophins.
Causes
• p hypogonadism:
• Testicular destruction/infiltration. Due to infection (CMV most
commonly, MAI, toxoplasmosis, TB) or neoplasm (lymphoma,
Kaposi’s sarcoma, germ cell tumours).
• Drug-induced. Ketoconazole (inhibits steroidogenesis, causing
lowered testosterone levels), megestrol acetate, other
glucocorticoids.
• s hypogonadism. Due to malnutrition, severe acute illness, destructive
disorders of pituitary/hypothalamus (CMV, toxoplasmosis, lymphoma);
medications, such as megestrol acetate (glucocorticoid-like action
causes hypogonadotrophic hypogonadism).
Females
• Hypogonadism in ♀, as evidenced by oligo-/amenorrhoea, less
common than in ♂ unless advanced disease. Fertility rates not affected
until advanced disease.
• Hypoandrogenism (testosterone, DHEA) common in ♀ with wasting
syndrome.
Electrolyte disturbance due to endocrine perturbation
in HIV/AIDS
Hyponatraemia
Very common in advanced disease. Due to SIADH in 50%; adrenal insuf-
ficiency also a common cause.
Calcium disorders
• Hypocalcaemia. Common (18% of patients with AIDS). Main cause is
vitamin D deficiency. Other causes: severe illness; hypomagnesaemia;
altered PTH secretion/metabolism; malabsorption of calcium and
vitamin D due to GIT opportunistic infection; medications (foscarnet
(complexes with calcium), pentamidine (induces renal magnesium
wasting and s PTH deficiency)).
• Hypercalcaemia. Rare. May relate to lymphoma or granulomatous disease.
656 CHAPTER 12 Other clinical/physiological situations
Thyroid
• Overt thyroid dysfunction is uncommon. Most common thyroid
dysfunction is sick euthyroid syndrome (non-thyroidal illness). i
thyroid-binding globulin often observed (significance unknown).
• Subclinical hypothyroidism may occur during HAART (highly acitve
antiretroviral therapy).
• Infections. Rare; usually post-mortem diagnoses. Thyroid function
usually euthyroid or sick euthyroid.
• Pneumocystis carinii (may also cause a thyroiditis).
• Mycobacteria.
• Cryptococcus neoformans.
• Aspergillosis.
• Neoplasm. Rare. Usually eu- or sick euthyroid; may be hypothyroid due
to infiltrative destruction.
• Kaposi’s sarcoma.
• Lymphoma.
Pituitary
• Anterior hypopituitarism. Very rare.
• Posterior pituitary dysfunction, causing diabetes insipidus (DI). Common.
• Infection. Toxoplasmosis, TB.
• Neoplasm. Cerebral lymphoma.
Wasting syndrome
Definition
The involuntary loss of >10% of baseline body weight, in combination
with diarrhoea, weakness, or fever. Wasting is an AIDS-defining condition.
Cause
Unknown but, in part, reflects d calorie intake due to anorexia associ-
ated with s infection. Underlying i resting energy expenditure associ-
ated with HIV infection per se. Hypogonadism common in ♂ with wasting
syndrome.
Treatment
• Highly active antiretroviral therapy (HAART). Associated with overall
weight gain, though lean body mass may remain unchanged.
• Nutritionally-based strategies. Adequate caloric intake to meet
metabolic demands. Efficacy limited, as refeeding generally increases fat
body mass, with little/less effect on lean body mass.
• Appetite stimulants. Megestrol acetate increases caloric intake and
weight compared to placebo, though most of weight gain due to i fat
mass. Dronabinol stimulates appetite, but weight gain is minimal.
• Exercise. Although exercise can increase total and lean body mass
in patients with AIDS, its role in patients with wasting syndrome is
not known.
• Androgen therapy. In hypogonadal ♂ patients with wasting syndrome,
testosterone increases overall weight and, in particular, lean body
mass. Both IM and transdermal testosterone effective. Testosterone
therapy not indicated in eugonadal ♂ with wasting.
ENDOCRINE DYSFUNCTION AND HIV/AIDS 657
Prolactin
Hyperprolactinaemia is common in ESRF but is usually mild, i.e.
<1,000mU/L. The cause is both i secretion and d renal clearance.
Gonadal function
• Hypogonadism—clinical and biochemical—is common in ESRF.
• In ♂, there is impaired pulsatile release of LH, although basal LH
levels are usually elevated due to impaired renal clearance. Serum FSH
is usually normal or mildly elevated.
• In ♀, levels of oestradiol, progesterone, and FSH are reported to
be within the normal range in the early follicular phase but fail to
show the usual cyclical changes. Menstrual disturbance is common.
Amenorrhoea, polymenorrhoea, and menorrhagia can also occur
on dialysis. Infertitility is the rule, and conception on dialysis is the
exception.
• Sexual dysfunction is common in both sexes but has been better
studied in ♂. 60% of ♂ have some degree of impotence, and
examination yields 80% to have testicular atrophy and 14% to have
gynaecomastia.
• Treatment of hypogonadism in ESRF is suboptimal. Testosterone
therapy is not associated with any clinical benefit in ♂.
Growth hormone and growth retardation
• Basal GH levels are normal, but there is impaired secretion following
an adequate hypoglycaemic stimulus in 40–70% of patients with ESRF.
• There is impaired growth in children, particularly during periods of
greatest growth velocity, and puberty is delayed. This combination
leads to short stature. The improved growth velocity after renal
transplantation is often too little too late in order to attain a normal
stature.
• Recombinant human GH (rhGH) has been shown to be an effective
treatment for growth retardation in children with stable chronic renal
failure (CRF) and ESRF as well as after renal transplantation.
Thyroid
The ‘sick euthyroid’ finding is common in CRF (b see Sick euthyroid
syndrome, p. 24).
Adrenal
• The adrenal axis is not impaired clinically by CRF.
• There is evidence of blunted cortisol response to hypoglycaemia, but
this is not relevant clinically.
• Patients with amyloidosis are at risk of hypoadrenalism due to adrenal
amyloid infiltration.
662 CHAPTER 12 Other clinical/physiological situations
Growth hormone
• In critical illness, the GH axis is profoundly affected, with initially raised
GH secretion but low IGF-1, IGFBPs, and GHBP related to peripheral
GH resistance.
• Prolonged critical illness >5–7 days results in low GH and a blunted
response to GHRH.
• Recombinant GH was proposed as a beneficial agent for critical
illness; however, the evidence is lacking, and there are reports of a
detrimental effect.
Hormone replacement and critical illness
There is no evidence that, other than insulin, hormonal supplementation
in the critically ill improves outcome.
Further reading
Elleger B, Debaveye Y, Van den Berghe G (2005). Endocrine interventions in the ICU. Eur J Intern
Med 16, 71–82.
Isidori AM, Kaltsas GA, Pozza C, et al. (2006). The ectopic adrenocorticotropin syndrome: clinical
features, diagnosis, management, and long-term follow-up. J Clin Endocrinol Metab 91, 371–7.
664 CHAPTER 12 Other clinical/physiological situations
Flushing
• Gonadal failure (with flushing and sweats).
• Drugs.
• Chlorpropamide.
• Nicotinic acid.
• Antioestrogens.
• LHRH agonists.
• Carcinoid (dry flushing—no sweats).
• Mastocytosis.
• Medullary thyroid cancer.
• Anaphylaxis.
• Pancreatic cell carcinoma.
• Phaeochromocytoma (more often pallor).
• Fever.
• Alcohol.
• Autonomic dysfunction.
• Some foods:
• Fish.
• Tyramine-containing food (cheese).
• Nitrites (cured meat).
• Monosodium glutamate.
• Spicy food.
• Gustatory flushing.
• Benign cutaneous flushing.
• Idiopathic.
Initial evaluation of patients with flushing
• Careful history.
• Physical examination (ideally during a flush although not often
possible):
• Examine skin carefully.
• Pulse rate.
• BP.
• Thyroid examination.
• Respiratory examination (wheeze).
• Careful abdominal examination.
• Urine dipstix.
• Biochemistry:
• Gonadotrophin levels.
• 2x 24h urinary 5HIAA measurements.
• Serum chromogranin A.
• 2x 24h urinary catecholamine measurements.
• Plasma metanephrines (if high level of suspicion).
• Serum tryptase level (if suspecting mastocytosis).
• Calcitonin level (if suspecting medullary thyroid cancer).
• Plasma VIP (if suspecting pancreatic carcinoma).
• Immunoglobulin levels (raised IgE may suggest allergies).
• Specific investigations, according to suspected diagnosis.
DIFFERENTIAL DIAGNOSIS OF POSSIBLE ENDOCRINE DISORDERS 669
Management of flushing
• Treat the underlying cause.
• Nadolol (non-selective B-blocker) effective in some cases of benign
cutaneous flushing.
• Somatostatin analogues can be used to treat flushing associated with
carcinoid syndrome.
• Antihistamines may be effective in some histamine-secreting carcinoid
tumours.
Further reading
Cleare A (2003). The neuroendocrinology of chronic fatigue syndrome. Endocr Rev 24, 236–52.
Cornuz J, Guessous I, Favrat B (2006). Fatigue: a practical approach to diagnosis in primary care.
CMAJ 174, 765–7.
Eisenach J, Atkinson J, Fealey R (2005). Hyperhidrosis: evolving therapies for a well established
phenomenon. Mayo Clin Proc 80, 657–66.
Izikson L, English J, Zirwas M (2006). The flushing patient: differential diagnosis, workup and
treatment. J Am Acad Derm 55, 193–208.
Paisley AN, Buckler HM (2010). Investigating secondary hyperhidrosis. BMJ 341, c4475.
670 CHAPTER 12 Other clinical/physiological situations
Metabolism
• GCs, via their direct effect and via reduced GH and sex hormone
activity, result in muscle and bone catabolism and fat anabolism.
• Chronic activation of the stress system is associated with i visceral
adiposity, d lean body mass, and suppressed osteoblastic activity
(which may ultimately lead to osteoporosis).
• GCs induce insulin resistance and other features of the metabolic
syndrome.
Other effects
• Immune: activation of the HPA axis inhibits the immune/inflammatory
response; most components of the immune response are inhibited by
glucocorticoids (>20% of genes expressed in human leukocytes are
regulated by glucocorticoids), increasing the susceptibility to infections.
Further reading
Charmandari E, Tsigos C, Chrousos G (2005). Endocrinology of the stress response. Annu Rev Physiol
67, 259–84.
672 CHAPTER 12 Other clinical/physiological situations
Endocrinology of exercise
Exercise and the hypothalamo–pituitary axis
Exercise presents a significant challenge to physiological homeostasis. The
endocrine system is integral to the body’s ability to adapt to exercise,
allowing the mobilization of metabolic fuels and also assisting in key cardi-
orespiratory responses.
• A ‘stress’ response is a significant part of this, with both CRH
and ADH stimulating the release of ACTH and hence cortisol.
This response promotes gluconeogenesis and helps to limit
exercise-induced inflammation. Chronically trained athletes
demonstrate a background hypercortisolaemia, although an attenuated
cortisol rise in response to exercise is also seen.
• GH is released in response to acute exercise and remains i until
around 2h afterwards, although the mechanisms underlying this are not
clear. The response is proportional to both the intensity and duration
of the exercise. In longer term, both 24h GH secretion and IGF-1
levels correlate well with physical activity and VO2max.
• In order that fluid homeostasis can be maintained, ADH is released
in response to exercise in response to osmotic stimuli. However,
non-osmotic stimuli can also affect ADH release, and this may
contribute to the development of exercise-associated hyponatraemia.
• Prolactin is secreted in response to an exercise bout, although regular
exercise does not seem to affect prolactin levels long-term. The role of
prolactin in exercise is not clear, but it may impact on immune function.
• Both prolactin and cortisol suppress GnRH secretion, and hence
LH/FSH secretion, which may explain why exercise can suppress
gonadal function. Potential effects of this are discussed in The female
athlete triad on b p.673.
• TSH is rapidly stimulated in response to acute exercise, but
longer-term effects are not clear. Exercise training can inhibit
peripheral thyroid hormone metabolism, leading to increased levels of
reverse T3 and increased levels of T3.
Exercise and bone health
• Physical activity has been shown to i bone mass, especially at
load-bearing sites. Approximately 26% of total adult bone mass is
gained in 2 years around the time of peak bone gain (12.5 years in
girls and 14.1 years in boys), meaning that physical activity may be
particularly important around this time.
• Bone strength may be a more important measure than bone mass.
Small, but significant, exercise-related i in bone strength have also
been seen in the lower extremities in children.
• In older adults, bone mass is improved with weight-bearing exercise,
compared with controls, in general due to the attenuation of normal
bone loss rather than an i in bone mass in the intervention group.
• Rates of stress fracture in athletes have been d with calcium and
vitamin D supplementation, although the evidence is not yet strong
enough for supplementation to be recommended routinely. Dosing is
also not clear.
ENDOCRINOLOGY OF EXERCISE 673
Tea
• Tea consists of green tea (unfermented), oolong tea (partially
fermented), and black tea (completely fermented).
• All teas contain fluoride and have high isoflavonoid content, in addition
to caffeine.
• Coffee (with a high caffeine content) has been associated with i hip
fracture risk. However, tea-drinking of all types has been associated
with higher BMD, although no fracture outcome has been reported.
DHEA
b see Miscellaneous alternative therapy p. 682.
Wild yam
Wild yam contains diosgenin which is used commercially as a source for
DHEA synthesis; however, this does not occur in humans.
Other compounds used by patients for osteoporosis
• Flaxseed (alpha-linolenic acid and lignans).
• Gelatin.
• Dong quai.
• Panax ginseng.
• Alfalfa.
• Liquorice.
There is no evidence of a +ve effect upon bone of these compounds.
682 CHAPTER 12 Other clinical/physiological situations
Diabetes
Classification
During the 1980s, the WHO published the first widely accepted classifica-
tion of diabetes. Diabetes was classified as insulin-dependent DM (IDDM)
or type 1 diabetes, and non-insulin-dependent DM (NIDDM) or type 2
diabetes. In 1997, the updated classification of diabetes retained the terms
type 1 and type 2 diabetes, discarding the terms IDDM and NIDDM. The
updated classification system focused on the specific underlying aetiology
of diabetes. These aetiological groups are listed in Box 13.1.
The vast majority of patients with diabetes have either type 1 diabe-
tes (secondary to autoimmune-mediated B-cell destruction and absolute
insulin deficiency) or type 2 diabetes (due to insulin resistance and defects
of insulin secretion). Table 13.2 outlines the key differences. Although the
remaining aetiological subtypes account for <10% of all diabetes cases, it
is important to consider these rarer causes of diabetes, which comprise
the entire differential diagnosis, in order to facilitate personalized manage-
ment of these patients. Rarer forms, such as maturity onset diabetes of the
young (MODY), largely arise in young adults. These individuals are often
assumed to have type 1 or type 2 diabetes and frequently experience long
delays before the correct diagnosis is reached. Fig. 13.1 illustrates a sug-
gested diagnostic algorithm for young adults.
Yes
Diabetes diagnosed ≤6 months
Genetic testing for neonatal diabetes
No
Yes
Yes
No
Consider MODY genetic testing
Genetics
Type 1 diabetes
The overall lifetime risk of developing type 1 diabetes in a Caucasian popu-
lation is currently 0.4%. This risk increases to:
• 1–2% if your mother has type 1 diabetes.
• 3–6% if your father has type 1 diabetes.
• 5–6% if a sibling has type 1 diabetes.
• Monozygotic twins have 750% concordance rate by age 40 years.
Early linkage studies identified the importance of the human leukocyte anti-
gen (HLA) genes of the major histocompatibility complex (MHC) in type 1
diabetes susceptibility. These variants account for half of the heritability of
type 1 diabetes. The HLA class II DR and DQ loci (respectively encoded
by genes DRB and DQB) account for almost all type 1 diabetes suscep-
tibility from this region. >90% of patients with type 1 diabetes carry at
least one copy of the DRB*301-DQB*201 or DRB*401-DQA*301-DQB*302
(allele numbers after asterisk).
Other susceptibility loci include variants in the gene encoding insulin
(INS) which confers a 2-fold increased risk. More recent genome-wide
association studies (GWAS) have increased the total number of loci asso-
ciated with type 1 diabetes to >40; however, these loci have much lower
effect size than the HLA or INS genes.
These genetic studies have confirmed that the pathogenesis of type 1
diabetes involves disordered immune regulation. Indeed, i levels of islet
cell antibodies (ICA), anti-glutamic acid decarboxylase (GAD) antibod-
ies, and anti-tyrosine phosphatase antibodies (anti-IA-2 antibodies) are
usually detected at diagnosis. The presence of all three antibodies give a
non-diabetic individual an 88% chance of developing type 1 diabetes over
the next 10 years.
Type 2 diabetes
The importance of genetic background in the aetiology of type 2 diabetes
is well established from family and twin studies (concordance between
monozygotic twins is 60–100%). Heritability of type 2 diabetes is estimated
at 725%.
• So far, >70 genetic susceptibility variants for type 2 diabetes have been
detected, largely through GWAS.
• TCF7L2 (encoding transcription factor 7-like 2 protein) in chromosome
10q is the susceptibility locus, with the largest effect on type 2 diabetes
risk, described to date, with a per-allele odds ratio of 71.4.
• Together, the genetic loci found so far only explain a small proportion
(710%) of the overall heritable risk for type 2 diabetes.
• Ongoing research is focused on identifying rare genetic variants which
might account for the missing heritability in type 2 diabetes.
• Potential areas of clinical translation include risk prediction,
prevention, pharmacogenetics, and development of novel
therapeutics.
GENETICS 691
Minor illness
KETONES urine –/+ OR blood <1.5mmol/L.
• If blood glucose >8mmol/L, increase all insulin doses by 10–20%.
• Check blood glucose and ketones every 4–6h.
• Take correction insulin (quick-acting insulin at usual correction
doses) every 4–6h, even if not eating.
Severe illness
KETONES urine ++/+++ OR blood >1.5mmol/L.
• Increase background insulin by 30–40%.
• KETONES 1.5–3mmol/L: take 10% of total daily insulin dose every 2h
as quick-acting insulin until ketones go away.
• KETONES >3mmol/L: take 20% of total daily insulin dose every 2h as
quick-acting insulin until ketones go away.
• If ketones do not fall after 2–4h, seek medical advice.
When ketones –ve/trace or <1.5mmol/L, carry on as for ‘minor illness’.
Try to eat 10–20g carbohydrate 4-hourly. Test frequently.
Calculation example
A patient has a standard regimen of basal insulin 12 units bd and 1
unit/10g CHO, giving an average TDD of 50 units.
• If unwell and hyperglycaemic but ketones <1.5mmol/L, increase basal
insulin to 13–14 units bd, and take regular correction doses.
• If blood ketones are 1.5–3mmol/L, take 10% of TDD = 5 units
2-hourly till ketone –ve.
• If blood ketones are >3mmol/L, take 10 units 2-hourly till
ketone –ve.
3 If ketones do not fall with these approaches and patients become more
ill or cannot take fluids due to nausea and vomiting, they should seek urgent
medical advice and will usually require admission.
SICK DAY RULES 701
702 CHAPTER 13 Diabetes
Hypoglycaemia
Management of hypoglycaemia (b see p. 746)
Hypoglycaemia (low blood glucose) is defined as blood glucose below
3.9 mmol/L, resulting from an imbalance between glucose supply, glucose
utilization, and current insulin levels. This is characterized by typical symp-
toms (see Table 13.3).
Autonomic symptoms are associated with a release of catecholamines,
cortisol, glucagon, and growth hormone. They typically occur at a blood
glucose level of around 3.8mmol/L while neuroglycopenic symptoms
occur when glucose levels drop below 3mmol/L. In the elderly or those
with long diabetes duration, autonomic symptoms are less pronounced
and occur at lower blood glucose levels, often resulting in delay in detect-
ing hypoglycaemia. This can also result in neuroglycopenia and confusion,
causing failure of recognition of hypoglycaemia.
Risk factors and causes of hypoglycaemia are listed in Box 13.4.
Severe hypoglycaemia
• This is defined as an episode that requires assistance from another
person, or that requires hospitalization or parenteral glucose
administration or treatment with glucagon.
• This occurs when blood glucose drops to a level inadequate to
support consciousness, usually below 1.5mmol/L.
• It is thought that, in some cases, this is associated with seizures or
cardiac arrhythmias and can be associated with death. The term ‘dead
in bed’ describes the scenario seen in about 6% of deaths in young
people with type 1 diabetes where the patient is found dead in an
undisturbed bed. There is often a history of recent susceptibility to
nocturnal hypoglycaemia.
Impaired awareness of hypoglycaemia
Recurrent hypoglycaemia can reduce the hormonal and symptomatic
responses to subsequent hypoglycaemia, and, over time, after exposure to
multiple episodes, patients can lose the ability to recognize hypoglycaemia.
This is termed hypoglycaemia unawareness. Those with impaired aware-
ness of hypoglycaemia have a 3–5-fold greater risk of severe hypoglycae-
mia, compared to those with normal awareness. Often, other people will
recognize hypoglycaemia before the patient does.
A simple score to assess awareness of hypoglycaemia is called the
GOLD score (see Fig. 13.2).
HYPOGLYCAEMIA 703
Ask patient: “How often are you aware of episodes of hypoglycaemia (cap-
illary blood glucose <3.5mmol/L)?”
Always Never
1 2 3 4 5 6 7
Using downloads
All available insulin pumps allow all their data to be downloaded to a
computer. This allows the user and the healthcare professional to analyse
the information and look for patterns that may be associated with either
hyper- or hypoglycaemia.
Bolus calculators
All currently available pumps have in-built bolus calculators. These can be
programmed with insulin-to-carbohydrate ratio, insulin sensitivity, and tar-
get blood glucose values. The calculator then takes into account the effect
of any previous boluses that were administered that may still be having a
glucose-lowering effect and provides a recommendation for a bolus. This
prevents ‘stacking’ of insulin and reduces the risk of hypoglycaemia.
Advanced pumping
Temporary basal rates
These allow the patient to increase or decrease the basal rate for a limited
duration of time. For example, to avoid the risk of hypoglycaemia during
exercise, the patient can reduce the basal rate to 50% of usual during
exercise. Similarly, if stressed or unwell and blood glucose is running high,
the patient can increase the basal rate by 70% to bring them under control.
Altered wave boluses (see Box 13.5)
Pumps offer the ability to deliver the bolus over different time periods to
cover different types of food.
Square wave bolus: this delivers the bolus over a given period of time
and is useful if someone plans to eat gradually over a given time period,
e.g. grazing on popcorn during a movie.
Use of downloads
Most modern home blood glucose meters and all currently available insu-
lin pumps have the capability to download data to a computer. These
data can then be used to obtain statistics, such as mean glucose, standard
deviation, and number of hypo- or hyperglycaemic excursions. They can
also be used to look for trends or patterns that can be used to inform
changes to therapy.
Pump downloads can be used to evaluate total daily dose, frequency of
set changes, and number of boluses/day and, when combined with data
from a meter, can provide very useful data on patterns.
Patients can download their pumps and/or meters and send the informa-
tion electronically to their diabetes team, allowing remote consultations
and making it easier for the diabetes team to provide dose adjustment
advice.
Suggested use of downloads
• Look for mean tests/day.
• Look for mean boluses/day.
• Look for mean total daily dose and basal/bolus split.
• Pattern management:
• Look at different time periods (overnight/post-breakfast/post-lunch
and post-evening meal), and try to identify patterns.
• Look for low blood glucose readings, and use the download
to identify possible causes (high insulin-to-carbohydrate ratio,
correction bolus, basal rate too high).
• Look for very high blood glucose, and look for possible causes
(missed bolus, inadequate insulin-to-carbohydrate ratio,
overtreatment of hypo).
710 CHAPTER 13 Diabetes
Emerging therapies
Closed-loop systems
The latest generation of sensor-augmented pumps can suspend insu-
lin delivery for up to 2h if the patient fails to respond to a hypoglycae-
mia alarm. This is particularly useful in preventing prolonged nocturnal
hypoglycaemia and has been shown to reduce the duration of nocturnal
hypoglycaemia in those most at risk. Currently, multiple groups are devel-
oping further generations of closed-loop systems, testing algorithms that
adjust insulin delivery through the pump, based on glucose data obtained
from CGM.
Immune modulation
Risk of type 1 diabetes can be predicted in siblings of patients with type 1
diabetes. Currently, immune modulation with anti-CD3 agents is in clini-
cal trials to try and stop the progression of the condition. To date, these
studies have produced small, but significant, protection of endogenous
insulin production and reduction in insulin doses, but the data do not cur-
rently justify clinical use outside of clinical trials. For more information on
trials on immune modulation in type 1 diabetes M http://www.bris.ac.uk/
trialnet-uk/index.html
ISLET CELL TRANSPLANTATION 711
Pancreas transplantation
Pancreas transplantation is most commonly performed simultaneously
with renal transplant in patients with type 1 diabetes and renal failure.
Occasionally, it may be performed alone for similar indications as islet
cell transplant in whom islet cell transplantation is considered inappro-
priate. This procedure has a 4% operative mortality, with 10–15% risk
of re-laparotomy. At 1 year, 95% are insulin-independent, with between
65 and 70% insulin-independent at 5 years. The outcomes are better for
simultaneous pancreas kidney transplant (SPK) than pancreas transplant
alone (PTA) or pancreas after kidney (PAK). These procedures should
be carried out at centres with expertise in transplantation and diabetes
management following multidisciplinary assessment.
Further reading
American Diabetes Association (2005). Defining and reporting hypoglycemia in diabetes: a report
from the American Diabetes Association Workgroup on Hypoglycemia. Diabetes Care 28,
1245–9.
DAFNE Study Group (2003). Training in flexible, intensive insulin management to enable dietary
freedom in people with type 1 diabetes: dose adjustment for normal eating (DAFNE) rand-
omized controlled trial. Diabet Med 20 Suppl 3, 4–5.
Hovorka R, et al. (2010). Manual closed-loop insulin delivery in children and adolescents with type
1 diabetes: a phase 2 randomised crossover trial. Lancet 375, 743–51.
JDRF CGM Study Group (2008). Continuous glucose monitoring and intensive treatment of type 1
diabetes. N Engl J Med 359, 1464–76.
National Institute for Health and Clinical Excellence (2004). NICE Guidelines, type 1 diabetes in
adults. Available at: M http://guidance.nice.org.uk/CG15/Guidance//Adults.
National Insitute for Health and Clinical Excellence (2008). NICE Guidelines, diabetes insulin
pump therapy. Available at: M http://publications.nice.org.uk/continuous-subcutaneous-insulin-
infusion-for-the-treatment-of-diabetes-mellitus-ta151/guidance.
Effect of intensive therapy on the development and progression of diabetic nephropathy in the
Diabetes Control and Complications Trial. The Diabetes Control and Complications (DCCT)
Research Group (1995). Kidney Int 47, 1703–20.
Hypoglycemia in the Diabetes Control and Complications Trial. The Diabetes Control and
Complications Trial Research Group (1997). Diabetes 46, 271–86.
Pickup J, Mattock M, Kerry S (2002). Glycaemic control with continuous subcutaneous insulin infu-
sion compared with intensive insulin injections in patients with type 1 diabetes: meta-analysis of
randomised controlled trials. BMJ 324, 705.
Shapiro AM, et al. (2006). International trial of the Edmonton protocol for islet transplantation.
N Engl J Med 355, 1318–30.
EXPERT MANAGEMENT OF TYPE 2 DIABETES 713
Side effects
Predominantly hypoglycaemia and weight gain. In the UKPDS, the mean
weight gain seen after 10 years of therapy was 2.3kg. Hypoglycaemia is
grossly under-reported with sulfonylurea therapies, with recent data sug-
gesting that up to 50% of patients experience at least one episode of low
blood sugar each year. The elderly are at particular risk. The longer-acting
agents, such as glibenclamide, should, if possible, be avoided.
Special precaution
Avoid in patients with porphyria.
Rapid-acting insulin secretagogues
These agents, whilst more commonly used in East Asian countries, are
not frequently prescribed in the UK. They include repaglinide, a carba-
moylmethyl benzoic acid derivative, and nateglinide, a D-phenylalanine
derivative, both of which stimulate insulin secretion with a short duration
of action. They are given 3 times a day and often referred to as prandial
glucose regulators. The short duration of action is associated with low
rates of hypoglycaemia, and they have been shown to reduce HbA1c by
0.6–2% (7–22mmol/mol). They should not be used in people with renal
and hepatic impairment.
716 CHAPTER 13 Diabetes
Thiazolidinediones (glitazones)
Pioglitazone is generally only used as an alternative to insulin in peo-
ple suboptimally controlled on metformin and/or sulfonylurea because
of its side effects. Its indication does, however, vary around the world,
and, in the UK, it is also considered in combination with insulin. The
recent introduction of incretin-based therapies (b see Oral and inject-
able incretin-based therapies, Table 13.5) appears to have limited its use
even further. A reduction of 0.6–1.5% (7–17mmol/mol) in HbA1c can be
expected, with low rates of hypoglycaemia.
Mode of action
Insulin-sensitizing agents by activating the peroxisome proliferator acti-
vated receptor (PPAR-G) which stimulates gene transcription for glucose
transporter molecules, such as Glut 1 and Glut 4.
Side effects
Patients should be warned about the possibility of significant oedema and
weight gain. Pioglitazone should not be used in people with evidence of
heart failure and those at a higher risk of bone fracture and osteoporosis.
In contrast to troglitazone, improvements in liver function, especially with
non-alcoholic steatohepatitis (NASH), have been reported.
Special precaution
Troglitazone was withdrawn soon after its UK launch because of reports
of hepatotoxicity. The second of these agents, rosiglitazone, whilst not
associated with hepatotoxicity, was associated with other significant side
effects, including ischaemic heart disease and increased cardiovascular
morbidity, and was withdrawn in 2011.
Alpha-glucosidase inhibitors
Acarbose is generally only considered in patients unable to tolerate other
oral glucose-lowering agents. When taken with food, it reduces postpran-
dial glucose peaks by inhibiting the digestive enzyme A-glucosidase which
normally breaks carbohydrates into their monosaccharide components,
thus retarding glucose uptake. The passage of undigested carbohydrates
into the large intestine results in bacterial breakdown and the develop-
ment of common unpleasant GI side effects.
Oral and injectable incretin-based therapies
(See Table 13.5 for comparison.)
GLP-1 receptor analogues
Exenatide and liraglutide are licensed for the use in people with type 2 dia-
betes, in combination with other oral therapies and basal insulin, and are
available as subcutaneous injections. They are associated with an HbA1c
reduction of 1–2% or 11–22mmol/mol and are particularly attractive as
glucose-lowering agents because of low rates of hypoglycaemia and a
weight loss over 6 months of 73–4kg.
ORAL GLUCOSE CONTROL THERAPIES 717
Mode of action
The incretin hormones glucagon-like polypeptide-1 (GLP-1) and gastric
intestinal polypeptide (GIP), released from the GI tract in response to
nutrient ingestion, stimulate insulin and suppress glucagon secretion in a
glucose-dependent manner. Their extrapancreatic effects include a slow-
ing of gastric emptying and central effects on the brain, leading to satiety
and reduced appetite.
Side effects
Mainly gastrointestinal, with generally mild and transient nausea, vomiting,
and diarrhoea. Headaches and dizziness are also reported. Increased risk
of hypoglycaemia when used with sulfonylureas.
Special precautions
Should not be used in people with a history of pancreatitis.
Dipeptidyl peptidase-4 (DPP-4) inhibitors
Oral therapies that are being used increasingly after metformin in many
patients. Sitagliptin has the broadest indication for use and can be used
with insulin. Lead to a 0.4–1.0% or 5–11mmol/mol reduction in HbA1c
over a 12-month period, with very low rates of hypoglycaemia, and are
weight-neutral (maybe even helping to reduce weight).
Mode of action
Inhibition of enzymic degradation of incretin hormones GLP-1 and GIP.
Side effects
Few major side effects reported. Skin rashes and nasopharyngitis.
Special precautions
Should not be used in people with history of pancreatitis. Liver function
test monitoring with vildagliptin.
718 CHAPTER 13 Diabetes
Insulin therapy
The majority of patients with type 2 diabetes will require insulin injections
at some point in their lives. Most guidelines recommend the addition of
a basal insulin when other therapies are unable to achieve or maintain
adequate glycaemic control. When using a basal insulin, existing oral thera-
pies should be continued. The use of a sulfonylurea should be reviewed
if hypoglycaemia occurs. Many patients initiated on a basal insulin will
require further intensification within 12 months to either a basal bolus
regimen or twice daily pre-mix.
Insulin can also be initiated as twice daily pre-mix insulin, and sulfonylu-
reas are usually discontinued. Insulin can be used with sitagliptin and piogl-
itazone, although the latter should be discontinued if clinically significant
fluid retention develops.
The newer analogue insulins offer certain advantages over the older
human and animal insulins. They are, however, more expensive, and many
national and international guidelines suggest that their use in type 2 diabe-
tes should be restricted to people with difficulties on human and animal
insulin, in particular, hypoglycaemia.
For different insulin preparations in the management of type 1 diabetes,
b see Box 13.3, p. 696.
BARIATRIC SURGERY 719
Bariatric surgery
Gastric bypass appears to have significant, and usually persistent, effects
on the metabolic abnormalities of type 2 diabetes, including glucose tol-
erance, dyslipidaemia, and hypertension. Further randomized controlled
trials with predefined metabolic entry criteria and detailed follow-up are
required. (For a full discussion of bariatric surgery, b see Chapter 15,
Obesity p. 862.)
720 CHAPTER 13 Diabetes
Insulin infusion
• Start a continuous FRIII via an infusion pump. 50 units human soluble
insulin (Actrapid®, Humulin S®), made up to 50mL with 0.9% NaCl
solution.
• Infuse at a fixed rate of 0.1 unit/kg/h (i.e. 7mL/h if weight is 70kg).
Ongoing management of DKA after the first 60min is covered in detail in
the National Guidelines, available at: M http://www.diabetologists-abcd.
org.uk/JBDS/JBDS_IP_DKA_Adults.pdf.
This involves continual monitoring and reassessment of the patient to
ensure that metabolic targets are being achieved (see Box 13.12), stabiliza-
tion and treatment of precipitating factors, and that other medical inter-
ventions or monitoring (e.g NG tube, catheter) are not required.
Practice points
Infusion of large volumes of normal saline may lead to hyperchloraemic
acidosis; however, this is not a cause of significant morbidity or prolonga-
tion of inpatient stay.
Convert back to SC insulin when biochemically stable (blood ketones
<0.3, pH >7.3) and the patient is ready to eat and drink.
Involve the specialist diabetes team at an early stage. It is commonly said
that DKA is usually a failure of self-management, so assessing the reasons
behind admission and re-education to prevent recurrence are vital.
DIABETIC HYPERGLYCAEMIC EMERGENCIES 737
738 CHAPTER 13 Diabetes
Perioperative management
of diabetes
Poor perioperative glycaemic control leads to poor outcomes in a vari-
ety of surgical specialities. There is detailed guidance available in the Joint
British Diabetes Societies guideline on the perioperative management of
adult patients with diabetes undergoing surgery or procedures (M http://
www.diabetologists-abcd.org.uk/JBDS/JBDS_IP_Surgery_Adults_Full.pdf).
There are several reasons why patients experience problems:
• Failure to identify patients with diabetes.
• Lack of institutional guidelines for management of diabetes.
• Poor knowledge of diabetes amongst staff delivering care.
• Complex polypharmacy and insulin-prescribing errors.
It is advocated that, whenever possible, the HbA1c should be below
69mmol/mol (8.5%) prior to surgery.
For most procedures where only one meal will be missed, hypogly-
caemic therapy can be adjusted, as outlined in Table 13.9 (OHA) and
Table 13.10 (insulin). The use of the VRIII (Table 13.8) should be limited to
those who will miss more than one consecutive meal, those who require
emergency surgery, and those for whom there was no time to optimize
their glycaemic control prior to surgery. The rate of insulin infusion is as
described in Table 13.8.
PERIOPERATIVE MANAGEMENT OF DIABETES 743
Management of hypoglycaemia
(b see also Type 1 diabetes, p. 702.)
Hypoglycaemia is the commonest side effect of insulin and sulfonylurea
treatment. Hypoglycaemia should be excluded in any person with diabe-
tes who is acutely unwell, drowsy, unconscious, unable to cooperate, or
presenting with aggressive behaviour or seizures. The hospital environ-
ment presents additional obstacles to the maintenance of good glycaemic
control and the avoidance of hypoglycaemia.
Management of hypoglycaemia
Algorithm A: adults who are conscious, orientated, and able to swallow
Give 15–20g quick-acting carbohydrate of the patient’s choice:
• 5–7 Dextrosol® tablets (or 4–5 Glucotabs®).
• 90–120mL of original Lucozade® (preferable in renal patients).
• 150mL non-diet cola (a small can).
• 5 Jelly babies or fruit pastilles or 10 Jelly beans.
• 3–4 heaped teaspoons of sugar dissolved in water*.
• 150–200 mL pure fruit juice*.
*Some centres advocate ONLY glucose for the treatment of hypos.
Repeat capillary blood glucose measurement 10–15min later. If blood glu-
cose is less than 4.0mmol/L, repeat step 1 up to three times.
If blood glucose remains less than 4.0mmol/L after 45min or three
cycles, consider 1mg of glucagon IM (remembering that this may be less
effective in patients prescribed sulfonylurea therapy) or IV 10% glucose
infusion at 100mL/h.
Once the blood glucose is above 4.0mmol/L and the patient has recov-
ered, give 15g long-acting carbohydrate:
• Two biscuits.
• One slice of bread.
• 200–300mL glass of milk (not soya).
• Normal meal if due (must contain carbohydrate).
DO NOT omit insulin injection if due (a dose review may be required).
Relative hypoglycaemia
Adults who have poor glycaemic control may start to experience symp-
toms of hypoglycaemia above 4.0mmol/L. Adults who are experiencing
symptoms, but have a blood glucose level greater than 4.0mmol/L, should
consume a small carbohydrate snack only (e.g. one medium banana or a
slice of bread). All adults with a blood glucose level less than 4.0mmol/L,
with or without symptoms of hypoglycaemia, should be treated as out-
lined in Management of hypoglycaemia in the section above.
Algorithm B: adults who are conscious but confused, disorientated,
unable to cooperate, or aggressive but are able to swallow
• If patient is unable to follow algorithm A but is able to swallow, give
either 1.5–2 tubes GlucoGel®/Dextrogel®, squeezed into the mouth
between the teeth and gums, or (if ineffective) give glucagon 1mg IM.
MANAGEMENT OF HYPOGLYCAEMIA 747
• Monitor blood glucose levels after 15min. If still less than 4.0mmol/L,
repeat steps 1 and 2 up to three times.
• If blood glucose level remains less than 4.0mmol/L after 45min, give IV
10% glucose infusion at 100mL/h.
• Once blood glucose is above 4.0mmol/L and the patient has
recovered, give a long-acting carbohydrate, as outlined in Management
of hypoglycaemia, see b p. 746.
• DO NOT omit insulin injection if due (a dose review may be
required).
• NB Patients given glucagon require a larger portion of long-acting
carbohydrate to replenish glycogen stores (double the suggested
amount above).
Algorithm C: adults who are unconscious and/or having seizures and/or
are very aggressive OR patients who are nil by mouth
• Assess patient.
• If the patient has an insulin infusion in situ, stop it immediately.
• The following two options are both appropriate:
• Glucagon 1mg IM (remembering that this may be less effective in
patients prescribed sulfonylurea therapy and may take up to 15min
to work).
• If IV access available, give 75mL of 20% glucose or 150mL of
10% glucose over 12–15min. Repeat capillary blood glucose
measurement 10min later. If blood glucose less than 4.0mmol/L,
repeat.
• Once blood glucose is greater than 4.0mmol/L and the patient has
recovered, give a long-acting carbohydrate, as outlined in Management
of hypoglycaemia, see b p. 746.
• DO NOT omit insulin injection if due (dose review may be required).
• If the patient was on IV insulin, continue to check blood glucose every
30min until it is above 3.5mmol/L, then restart IV insulin after review
of dose regimen.
748 CHAPTER 13 Diabetes
Diagnosis of CVA
Fig. 13.3 Algorithm for managing diabetes in patient with a stroke and swallowing
difficulties.
THE MANAGEMENT OF STROKE PATIENTS WITH DIABETES 749
750 CHAPTER 13 Diabetes
Maternal
• Increased risk of severe hypoglycaemia (SH), particularly during early
pregnancy. Historically, the prevalence of SH was as high as 30–40% in
type 1 diabetes but may now be declining with increased use of insulin
analogues and pump therapy.
• Increased risk of pre-eclampsia, which is related to glycaemic control
and complicates approximately 15% diabetic pregnancies.
• Worsening of diabetic nephropathy and progression of
retinopathy: renal and retinal screening is recommended before
pregnancy and during each trimester.
• Increased risk of diabetic ketoacidosis (DKA). Although rare, affecting
<1% of pregnancies, DKA is associated with fetal loss in up to 20% of
episodes. Women with suspected DKA should be admitted to level 2
critical care for immediate medical and obstetric care.
• Increased delivery by Caesarean section (66% in type 1 diabetes).
• Thromboembolic disease, while rare, is responsible for a third of all
maternal deaths. All women with risk factors (age >35, BMI >30kg/m2,
proteinuria >5g/day) and/or other comorbidities should receive
prophylactic low molecular weight heparin throughout pregnancy.
• Thyroid dysfunction is three times more common in type 1 diabetes
pregnancy and should be assessed during pregnancy and post-partum.
Type 1 and type 2 diabetes
• Most women (95%) deliver normal healthy babies. The congenital
malformation and spontaneous abortion rates are higher when the
HbA1c is elevated (>6.1%; 43mmol/mol).
• Pregnancy is not recommended in women with HbA1c >86mmol/
mol (10%).
754 CHAPTER 13 Diabetes
Gestational diabetes
Epidemiology
Pregnancy induces a state of insulin resistance, with increases in the lev-
els of growth hormone, progesterone, placental lactogen, and cortisol,
resulting in impaired glucose disposal. Hyperglycaemia during pregnancy
occurs in up to 2–5% of women and is associated with increased risk of
subsequent type 2 diabetes in up to 50% women over the next 5–10 years.
The risk of subsequent type 2 diabetes is significantly reduced by diet and
lifestyle and by metformin.
There is a clear association with increasing hyperglycaemia and poorer
maternal and fetal outcomes. Intensive treatment with diet, metformin,
and insulin (required in 10–20% women) reduces the risk of serious
perinatal morbidity (death, macrosomia with associated complications of
shoulder dystocia, bone fracture, and nerve palsy), Caesarean delivery,
and maternal hypertensive disorders.
Diagnostic criteria for GDM are listed in Table 13.11.
High-risk groups
Most women with gestational diabetes are detected on routine screening
at 24–28 weeks, but certain high-risk groups should be screened earlier.
These risk factors include:
• Previous gestational diabetes.
• A large baby in their last pregnancy, e.g. >4.5kg.
• A previous unexplained stillbirth/perinatal death.
• Maternal obesity (BMI above 30kg/m2).
• Family history of diabetes (first-degree relatives).
• Family origin with a high prevalence of type 2 diabetes:
• South Asian.
• Black Caribbean.
• Middle Eastern.
• Polyhydramnios.
Screening
Screening is recommended at 16 weeks if previous gestational diabetes
and, if normal, should be repeated at 24–28 weeks in high-risk groups.
Box 13.15 details methods of screening.
Treatment
• Initial treatment is with dietary advice and exercise. There is an
emphasis on watching the quantity of carbohydrate (especially
at breakfast) and choosing carbohydrates from low glycaemic
index sources, lean proteins, including oily fish, and a balance of
polyunsaturated fats and monounsaturated fats.
• 30min of daily physical activity is recommended.
• Women with pre-pregnancy BMI ≥27kg/m2 should also be advised to
restrict calorie intake (to 25kcal/kg/day or less).
Oral hypoglycaemic therapy
Oral hypoglycaemic treatment is recommended if diet and exercise fail to
maintain blood glucose levels in the target range (3.5–7.8mmol/L) and/or
the growth scans suggest incipient macrosomia (abdominal circumference
>70th percentile).
Metformin and/or glibenclamide are not licensed for use in pregnancy
but are commonly used. Informed consent should be obtained.
Insulin therapy
• Required in 10–20% gestational diabetes pregnancies to maintain
fasting blood glucose 3.5–5.9mmol/L and 1h postprandial <7.8mmol/L.
• Used in conjunction with diet and exercise or in addition to
metformin.
• Regimen should be tailored to glycaemic profile and patient
acceptability: boluses alone, basal alone, mixed or basal bolus.
• Most (but not all) women can stop insulin and/or oral hypoglycaemic
treatments immediately after birth.
Post-partum follow-up
• Women with gestational diabetes should be offered lifestyle advice
(including weight control, diet, and exercise) and a fasting plasma
glucose measurement at the 6-week post-natal check and annually
thereafter.
• NICE do not recommend a post-partum OGTT, but this is often used
in high-risk multiethnic groups at increased risk of type 2 diabetes.
See Box 13.16 for a checklist for GDM during clinic visit.
758 CHAPTER 13 Diabetes
Further reading
Crowther CA, et al. (2005). Effect of treatment of gestational diabetes mellitus on pregnancy
outcomes. N Engl J Med 352, 2477–86.
International Association of Diabetes and Pregnancy Study Groups Consensus Panel (2010).
International Association of Diabetes and Pregnancy Study Groups recommendations on the
diagnosis and classification of hyperglycemia in pregnancy. Diabetes Care 33, 676–82.
Landon MB, et al. (2009). A multicenter, randomized trial of treatment for mild gestational diabetes.
N Engl J Med 361, 1339–48.
Macintosh MC, et al. (2006). Perinatal mortality and congenital anomalies in babies of women with
type 1 or type 2 diabetes in England, Wales, and Northern Ireland: population based study.
BMJ 333, 177.
Metzger BE, et al. (2008). Hyperglycemia and adverse pregnancy outcomes. N Engl J Med 358,
1991–2002.
National Institute for Health and Clinical Excellence (2008). NICE guideline 63: Diabetes in Pregnancy:
Management of diabetes and its complications in pregnancy from the pre-conception to the post-
natal period. Available at: M http://publications.nice.org.uk/diabetes-in-pregnancy-cg63/guidance.
Ratner RE, et al. (2008). Prevention of diabetes in women with a history of gestational diabetes:
effects of metformin and lifestyle interventions. J Clin Endocrinol Metab 93, 4774–9.
Rowan JA, et al. (2008). Metformin versus insulin for the treatment of gestational diabetes. N Engl
J Med 358, 2003–15.
GESTATIONAL DIABETES 759
760 CHAPTER 13 Diabetes
Management of children
with diabetes
In the UK, standards for best practice in caring for children with diabetes
have been linked to payment of a ‘best practice tariff’—a set amount of
money paid for each young person cared for (up to their 19th birthday).
To qualify for this, the following must apply:
• Every child or young person with diabetes will be cared for by a
specialist team of healthcare professionals (consisting of a doctor,
a nurse, and a dietician as a minimum) who have specific training in
paediatric diabetes. DSN caseload 70–100 patients.
• For new diagnoses: the case must be discussed with the specialist team
within 24h of the diagnosis and seen on the next working day.
• An age-appropriate structured education programme offered at
diagnosis and with updates, as needed.
• Four clinic appointments with HbA1c check.
• Annual dietician contact.
• At least eight additional contacts per year from members of the
specialist team (not all face-to-face) are recommended.
• Annual review with BP and retinopathy/renal screening from age 12.
• Annual psychology assessment and access to psychology services.
• 24h emergency phone advice for families/other health professionals.
• Involvement in the National Paediatric Diabetes Audit and Paediatric
Diabetes Network.
• Policy for transition to adult services.
• Policy on managing high HbA1c and infrequent clinic attendance.
Type 1 diabetes in children
Insulin
Insulin is started at a total daily dose of 0.5 units/kg body weight in children.
In puberty, the starting dose is 0.7 units/kg body weight.
Insulin regimens are the same as in adults, varying from centre to centre,
with very little evidence base for one being superior. In toddlers, basal
insulin tends to be given in the morning, moving to the evening in children
aged >5 years. It is often used twice daily in puberty. Prandial insulin doses
for food can be anything from 0.5 units for 40g of CHO in babies to 3–4
units for 10g in pubertal boys.
Continuous subcutaneous insulin infusion (CSII) pumps (b p. 704)
NICE guidelines allow for pumps to be funded in children under the age
of 12 in whom multiple insulin injections are considered to be impractical
or inappropriate. Over 12s have the same funding restrictions as adults.
Education
As in adults, this is the mainstay of management to allow parents and then
children to self-manage their own condition.
• Carbohydrate counting. This may be taught at any stage but is better if
done soon after diagnosis so that it becomes routine for the family.
• Multiple carers. One of the main differences between children and
adults with diabetes is the need to teach many family members about
the condition, including grandparents and childminders.
MANAGEMENT OF CHILDREN WITH DIABETES 765
• Blood testing. Before every meal and at other times to work out
whether mealtime doses are correct, as well as at other times for
troubleshooting. Many children require 6–8 blood tests per day, and
so there is much interest currently in continuous subcutaneous glucose
sensing.
• Exercise. Children’s exercise levels vary enormously from one day to
the next. Therefore, advice about how to cope with the BG variability
is essential and built into most structured education programmes.
• Management at school and nursery:
• Children spend around a third to half of waking hours at school.
• Schools need to support intensive management of diabetes.
• This works best when volunteers at primary school are taught
diabetes management. This includes either supervising or doing the
BG testing, insulin injections, or pump boluses, and, in some places,
the use of algorithms to calculate insulin doses for food and BG
correction. This involves a lot of education, usually by the DSN.
• All school staff (primary and secondary) should be aware of the
symptoms of hypoglycaemia in a particular child and should know
how to treat hypoglycaemia.
Acute complications are mainly treated as in adults (b see Expert man-
agement of type 1 diabetes, p. 694; Diabetes and life, p. 722; Hospital
inpatient diabetes management and diabetic emergencies, p. 732). Specific
points relevant to children are described in the following sections.
Hypoglycaemia
• Parents and carers should look out for symptoms, as children may not
recognize them reliably until around 10 years old.
• Dose of glucagon for severe lows with fits or unconsciousness: 0.5mg
if under 8 years old and 1.0mg if older.
Diabetic ketoacidosis
Management in those <18 years should be according to the British Society
of Paediatric Endocrinology and Diabetes guidelines (M http://www.
bsped.org.uk/clinical/clinical_endorsedguidelines.html). Key features which
are different from the new adult guidelines are:
• Lower volumes of fluid than in adults because of the risk of cerebral
oedema.
• Fixed-rate insulin infusion at 0.1 units/kg/h.
• Delay in insulin administration until after 1h of fluids for the same reason.
Complications and associated conditions
Annual review should include:
• Blood pressure at all ages.
• Microalbuminuria and retinopathy screening: annually when >12 years.
• Examine feet to instil good practices.
• 3-yearly screening for coeliac disease (6–8% patients have condition).
• Annual thyroid function tests.
• Addison’s disease—no screening, but consider if recurrent
hypoglycaemia and lethargy.
766 CHAPTER 13 Diabetes
• Smoking.
• Recreational drugs.
• Contraception and pre-conception care.
• Sick day rules—to take account of living alone for the first time.
• Eating disorders:
• Mild forms are common, especially, but not exclusively, in girls.
Discussion should be towards helping the young person recognize
the link between thoughts, feelings, and behaviour and to try to find
alternative behaviours. Missed insulin injections are often a way of
controlling weight.
• Higher education:
• Often secondary specialist care is kept at home for appointments
during the holidays, with local GP registration with the university/
college student health system. Young people on insulin need a
fridge to be provided in their University accommodation and to let
people on their corridor know about diabetes. A discussion about
how they will manage ‘fresher’s week’, especially with alcohol, and
other issues is helpful.
• Separation from parents:
• The paediatric team will usually have spent many years
communicating more with the parents than the children. However,
now the young person may need to be helped to understand their
diabetes for themselves, often requiring a new process of education
from basics onwards.
768 CHAPTER 13 Diabetes
Fig. 13.4 This photograph shows new vessels at the disc (NVD) and haemorrhage
from these new vessels in front of the inferior temporal arcade.
772 CHAPTER 13 Diabetes
New vessels on the disc (NVD) are defined as any new vessel devel-
oping at the optic disc or within 1 disc diameter of the edge of the
optic disc.
New vessels elsewhere (NVE) are defined as any new vessel developing
more than 1 disc diameter away from the edge of the optic disc.
Both give no symptoms but cause the problems of advanced retinopa-
thy, such as haemorrhage, scar tissue formation, traction on the retina, and
retinal detachment which actually results in loss of vision.
The Diabetic Retinopathy Study (DRS) recommended prompt treat-
ment with panretinal photocoagulation in the presence of DRS high-risk
characteristics, which reduced the 2-year risk of severe visual loss by 50%
or more and were defined by:
• The presence of pre-retinal or vitreous haemorrhage.
• Eyes with NVD equalling or exceeding 1/4 to 1/3 disc area in extent
with no haemorrhage.
• NVE equalling >1/2 disc area with haemorrhage (from the NVE).
Untreated, eyes with high-risk characteristics had between 25.6% and
36.9% chance of severe visual loss within 2 years.
Proliferative eyes without high-risk characteristics had the following
risks of severe visual loss:
• Untreated—2 years 7.0%, 4 years 20.9%.
• Treated—2 years 3.2% 4 years 7.4%.
Because the side effects of modern laser treatment are considerably less
than the early lasers, most ophthalmologists treat eyes that develop new
vessels of either the high- or low-risk categories.
Diabetic macular oedema
Diabetic macular oedema or maculopathy may be classified into focal,
diffuse, and ischaemic types.
In focal maculopathy, focal leakage tends to occur from microaneu-
rysms, often with a circinate pattern of exudates around the focal leakage.
In the diffuse variety, there is a generalized breakdown of the blood–
retina barrier and profuse early leakage from the entire capillary bed of
the posterior pole, sometimes accompanied by cystoid macular changes.
In ischaemic maculopathy, enlargement of the foveal avascular zone
(FAZ) due to capillary closure is found with variable degrees of visual loss.
The Early Treatment Diabetic Retinopathy Study (ETDRS) reported
that focal photocoagulation of ‘clinically significant’ diabetic macular
oedema (CSMO) substantially reduced the risk of visual loss, CSMO being
defined as:
• Thickening of the retina at or within 500 microns of the centre of the
macula.
• Hard exudates at or within 500 microns of the centre of the fovea,
if associated with thickening of the adjacent retina (no residual hard
exudates remaining after disappearance of retinal thickening).
• A zone or zones of retinal thickening 1 disc area or larger, any part of
which is within 1 disc diameter of the centre of the macula.
CLINICAL & HISTOLOGICAL FEATURES OF DIABETIC EYE DISEASE 773
774 CHAPTER 13 Diabetes
Eye screening
National screening programmes
The development of screening in Europe was first encouraged by the
St. Vincent Declaration which, in 1989, set a target for the reduction of
new blindness by one-third in the following 5 years.
National screening programmes, with consensus grading protocols,
were introduced in the UK in 2002–2004. The National Institute for
Health and Clinical Excellence (NICE) recommends that those with type 1
and type 2 diabetes have their eyes screened at the time of diagnosis and
at least annually thereafter. There are differences in the protocols used
globally in screening programmes. In England, the method used is two-field
mydriatic digital photography, with screening performed by technician
screeners or optometrists. Fixed locations and mobile units are both used.
Monitoring of programme performance is via Quality Assurance Standards
and Key Performance Indicators against which individual screening pro-
grammes are assessed.
Ad hoc eye examinations
It is important to remember that the patients who are most at risk of
developing sight-threatening diabetic retinopathy and visual loss are the
regular non-attenders for screening. Hence, it is important to examine
the eyes of patients who have not attended for routine screening using
the following method:
Visual acuity
Use a standard Snellen chart for distance, and check each eye separately.
Let the patient wear their glasses for the test, and if vision is worse
than 6/9, also check with a pinhole, as this will correct for any refractive
(glasses) error. If it does not correct to 6/9 or better, consider more
careful review; some maculopathy changes cannot be seen easily with a
handheld ophthalmoscope, and an ophthalmology review may be needed.
Cataracts are a more likely cause, so look carefully at the red reflex.
High blood glucose readings can lead to myopia and low blood glucose
to hypermetropia. In both these circumstances, one would expect the
vision to improve with a pinhole.
Eye examination
• Dilate the pupil before looking into the eye.
• Use tropicamide 1%—dilates the pupil adequately in 15–20min and
lasts only 2–3h.
• In those with a dark iris, you may also need phenylephrine (2.5%),
added soon after the tropicamide, to give adequate views.
• The main reasons not to dilate are closed angle glaucoma and
recent eye surgery, but as such patients are usually under an eye
clinic already, most people are suitable for dilatation.
• Once the pupil is dilated, look at the red reflex to check for lens
opacities. Examine the anterior chamber, as, although rare, rubeosis
iridis is important to pick up. The vitreous is examined before
EYE SCREENING 775
examining the retina. When examining the retina, use the optic disc
as a landmark; follow all four arcades of vessels out from it; examine
the periphery, and at the end, examine the macula, as this can be
uncomfortably bright through a dilated pupil, and if done at the start, it
makes it difficult for anyone to keep their eye still enough to complete
the examination adequately. It is usually much easier for the patient
to look directly at the light if a smaller spot size or a target on a green
background are chosen for this aspect of the examination.
When to refer
Referral will depend on local preferences but are based on NSC manage-
ment guidelines, as outlined in Box 13.18.
Further reading
Elman MJ, Aiello LP, Beck RW, et al. (2010). Randomized trial evaluating ranibizumab plus prompt
or deferred laser or triamcinolone plus prompt laser for diabetic macular edema. Ophthalmology
117, 1064–77.
Keech AC, Mitchell P, Summanen PA, et al. (2007). Effect of fenofibrate on the need for laser treat-
ment for diabetic retinopathy (FIELD study): a randomised controlled trial. Lancet 370, 1687–97.
Klein R, Knudtson MD, Lee KE, Gangnon R, Klein BE (2008). The Wisconsin Epidemiologic Study
of Diabetic Retinopathy: XXII the twenty-five-year progression of retinopathy in persons with
type 1 diabetes. Ophthalmology 115, 1859–68.
National Institute for Health and Clinical Excellence (2013). NICE appraisal: Ranibizumab for the
treatment of diabetic macular oedema. Available at: M http://www.nice.org.uk/nicemedia/
live/14082/62873/62873.pdf.
No authors listed (1985). Photocoagulation for diabetic macular edema. Early Treatment Diabetic
Retinopathy Study report number 1. Early Treatment Diabetic Retinopathy Study research
group. Arch Ophthalmol 103, 1796–806.
No authors listed (1987). Indications for photocoagulation treatment of diabetic retinopa-
thy: Diabetic Retinopathy Study Report no. 14. The Diabetic Retinopathy Study Research
Group. Int Ophthalmol Clin 27, 239–53.
No authors listed (1991). Grading diabetic retinopathy from stereoscopic color fundus
photographs--an extension of the modified Airlie House classification. ETDRS report number
10. Early Treatment Diabetic Retinopathy Study Research Group. Ophthalmology 98(5 Suppl),
786–806.
Raymond NT, Varadhan L, Reynold DR, et al. (2009). Higher prevalence of retinopathy in dia-
betic patients of South Asian ethnicity compared with white Europeans in the community: a
cross-sectional study. Diabetes Care 32, 410–15.
Wong TY, Mwamburi M, Klein R, et al. (2009). Rates of progression in diabetic retinopathy during
different time periods: a systematic review and meta-analysis. Diabetes Care 32, 2307–13.
Yau JW, Rogers SL, Kawasaki R, et al. (2012). Global prevalence and major risk factors of diabetic
retinopathy. Diabetes Care 35, 556–64.
SURGICAL TREATMENT FOR DIABETIC EYE DISEASE 781
782 CHAPTER 13 Diabetes
Blood pressure
Blood pressure is normal, or even low, at the onset of type 1 diabetes.
It increases with albuminuria such that over 50% of patients are on anti-
hypertensive therapy or have values >130/80mmHg by the time they
develop persistent microalbuminuria. In contrast, over 35% of newly diag-
nosed patients with type 2 diabetes in the UKPDS had a blood pressure
>160/90mmHg or were on treatment. More than 80% of patients with
overt nephropathy will be on antihypertensive therapy, irrespective of
their type of diabetes. Thus, blood pressure increases are a concomitant
factor of nephropathy in type 1 patients but may be more of a causa-
tive factor in type 2 diabetes. What is certain is that high blood pressure
drives nephropathy progression (increases in albuminuria and rate of loss
of GFR), and its management is critical in the prevention of ESRD.
Cardiovascular disease
Patients with nephropathy have a greatly increased risk of cardiovascular
disease partly because of increases in blood pressure, but also partly due
to increased insulin resistance and serum lipid abnormalities. Mortality
is increased at least 2-fold for patients with microalbuminuria. Recent
data in people with type 1 diabetes from Finland suggest that mortality
is increased only in those with increased albuminuria. The UKPDS cohort
had an annual mortality of 2–4% for those with microalbuminuria, but this
increased to nearly 20% in those with a serum creatinine >175micromol/L
and/or renal replacement therapy (RRT). This explains why many patients
fail to survive to RRT. Intensive management of modifiable cardiovascular
risk factors has been shown to improve both morbidity and survival in
people with type 2 diabetes and microalbuminuria, and the SHARP trial
has established the effectiveness of lipid-lowering therapy in preventing
cardiovascular events in patients with CKD stage 3 or worse.
788 CHAPTER 13 Diabetes
Further reading
Bilous R (2008). Microvascular disease: what does the UKPDS tell us about diabetic nephropathy?
Diabet Med Suppl 2, 25–9.
Bilous R, Chaturvedi N, Sjolie AK, et al. (2009). Effect of candesartan on microalbuminuria and
albumin excretion rate in diabetes: 3 randomised trials. Ann Intern Med 151, 11–20.
DCCT/EDIC Research Group (2003). Sustained effect of intensive treatment of type 1 diabetes
mellitus on development and progression of diabetic nephropathy. JAMA 290, 2159–67.
De Boer I, Rue T C, Cleary PA, et al. (2011). Long-term outcomes of patients with type 1 diabetes
mellitus and microalbuminuria. Arch Int Med 171, 412–20.
Forbes JM, Coughlan MT, Cooper ME (2008). Oxidative stress as a major culprit in kidney disease
in diabetes. Diabetes 57, 1446–54.
Gaede P, Lund-Andersen H, Parving H-H, Pedersen O (2008). Effect of multifactorial interventions
on mortality in type 2 diabetes. N Engl J Med 358, 580–91.
Joint Specialty Committee on Renal Medicine of the Royal College of Physicians and the Renal
Association, and the Royal College of General Practitioners (2006). Chronic Kidney Disease
in Adults: UK Guidelines for identification, management and referral. London: Royal College
of Physicians.
KDOQI (2007). KDOQI Clinical Practice Guidelines in Clinical Practice Recommendations for
Diabetes and Chronic Kidney Disease. Am J Kidney Dis 49(Suppl 2), S1–180.
Levey AS, Coresh J (2012). Chronic kidney disease. Lancet 379, 165–80.
National Collaborating Centre for Chronic Conditions (2008). Clinical guidance 73. Chronic Kidney
Disease. Early identification and management of chronic kidney disease in adults in primary and
secondary care. Available at: M http://guidance.nice.org.uk/CG73/QuickRefGuide/pdf/English.
Prigent A (2008). Monitoring renal function and limitation of renal function tests. Semin Nucl Med
38, 32–46.
TREATMENT FOR DIABETIC RENAL DISEASE 791
792 CHAPTER 13 Diabetes
Diabetic neuropathy
Definition
Involvement of cranial, peripheral, and autonomic nerves may be found in
patients with diabetes and termed diabetic neuropathy; this usually sug-
gests a diffuse, predominantly sensory peripheral neuropathy. The effects
on nerve function can be both acute or chronic as well as being transient
or permanent. The commonest clinical consequences of neuropathy are:
• Altered sensation (both pain and i sensitivity to normal sensation).
• Neuropathic ulcers, usually on the feet.
• Erectile dysfunction (with autonomic neuropathy).
• Charcot arthropathy.
See Box 13.22 for classification.
Pathology
Diabetic neuropathy is one of the microvascular complications of diabetes.
Pathologically, distal axonal loss occurs with focal demyelination and
attempts at nerve regeneration. The vasa nervorum often shows base-
ment membrane thickening, endothelial cell changes, and some occlusion
of its lumen. This results in slowing of nerve conduction velocities or a
complete loss of nerve function. Both metabolic and vascular changes have
been implicated in its aetiology.
Pathogenesis
Hyperglycaemia is probably the major underlying cause of the histological
and functional changes, although vascular risk factors, such as hypertension
and hyperlipidaemia, may also have a role. Several possible mechanisms
have been suggested:
• Overloading of the normal pathways for glucose metabolism, resulting
in i use of the polyol pathway which leads to i levels of sorbitol
and fructose and d levels of myoinositol and glutathione. This may
result in more free radical damage and also lowers nitric oxide levels,
thus altering nerve blood flow. Experimental models, using aldose
reductase inhibitors which can improve aspects of diabetic neuropathy,
support this theory.
• Accumulation of AGE (via non-enzymatic glycation) may contribute.
• In the more acute neuropathies, acute ischaemia of the nerves due to
vascular abnormalities has been suggested as the cause. The underlying
reason for this is still unclear. Insulin-induced ‘neuritis’ may occur when
insulin therapy is started and blood glucose levels fall.
• Other potential aetiological factors include changes in local growth
factor production and oxidative stress.
Further work is needed to clarify the exact role of each of the above
mechanisms. In the meantime, studies showing improvements in neuropa-
thy associated with good diabetic control strengthen the argument for the
role of hyperglycaemia and offer us a treatment option.
DIABETIC NEUROPATHY 793
Mononeuropathies
Peripheral mononeuropathies and cranial mononeuropathies are not
uncommon. These may be spontaneous or may be due to entrapment
or external pressure. Of the peripheral mononeuropathies, median nerve
involvement and carpal tunnel syndrome may be found in up to 10% of
patients and require nerve conduction studies and then surgical decom-
pression. Entrapment of the lateral cutaneous nerve of the thigh is also
seen more commonly in those with diabetes, giving pain over the lateral
aspect of the thigh. Common peroneal nerve involvement, causing foot
drop and tarsal tunnel syndrome, is also recognized but less common.
Cranial mononeuropathies usually occur suddenly and have a good
prognosis. Palsies of cranial nerves III and VI are most commonly seen
(but still infrequent). In the IIIrd nerve palsy, sparing of the pupillary
responses is usual. Spontaneous recovery is slow over several months,
and no treatment, apart from symptomatic help such as an eye patch, is
needed. Unlike entrapment neuropathies where decompression may help,
no effective treatment is currently available in most cases of spontaneous
mononeuropathy.
796 CHAPTER 13 Diabetes
Autonomic neuropathy
The commonest effect of autonomic neuropathy is erectile dysfunction
which affects 40% of ♂ with diabetes (although this is multifactorial).
The recent interest in sildenafil has highlighted this. Only a small number
develop severe GI, cardiac, or bladder dysfunction. Abnormal autonomic
function tests can be expected in 20–40% of a general diabetic clinic popu-
lation. The i problems during surgery from cardiac involvement should
be remembered.
Clinical features
• Erectile dysfunction.
• Postural hypotension—giving dizziness and syncope in up to 12%.
• Resting tachycardia or fixed heart rate/loss of sinus arrhythmia—in up
to 20%.
• Gustatory sweating—sweating after tasting food.
• Delayed gastric emptying, nausea/vomiting, abdominal fullness.
• Constipation/diarrhoea.
• Urinary retention/overflow incontinence.
• Anhidrosis—absent sweating on the feet is especially problematic, as it
increases the risk of ulceration.
• Abnormal pupillary reflexes.
Assessment
If AN is suspected, check:
• Lying and standing BP (measure systolic BP after 2min standing; normal
is <10mmHg drop; >30mmHg is abnormal).
• Pupillary responses to light.
Other less commonly performed tests to consider if the diagnosis is
uncertain or in high-risk patients include:
• Loss of sinus arrhythmia. Measure inspiratory and expiratory heart rates
after 5s of each (<10 beats/min difference is abnormal; >15 is normal).
• Loss of heart rate response to Valsalva manoeuvre. Look at the ratio
of the shortest R–R interval during forced expiration against a
closed glottis, compared to the longest R–R interval after it (<1.2 is
abnormal).
• BP response to sustained hand grip. Diastolic BP prior to the test is
compared to diastolic BP after 5min of sustaining a grip equivalent
to 30% of maximal grip. A diastolic BP rise >16mmHg is normal;
<10mmHg is abnormal. A rolled-up BP cuff to achieve the required
hand grip may be used.
• For gastroparesis, consider a radioisotope test meal to look for
delayed gastric emptying. Blood glucose should be <10mmol/L, as
hyperglycaemia exacerbates delayed gastric emptying.
Treatment
This is based on the specific symptom and is usually symptomatic only.
In all patients, improvement in diabetic control is advocated in case any
of it is reversible, but this is not usually very helpful or effective.
AUTONOMIC NEUROPATHY 801
Postural hypotension
Stopping drugs that may result in, or exacerbate, postural hypotension,
including diuretics, B-blockers, anti-anginal agents, tricyclic agents, etc.
• Advising patients to get up from the sitting or lying position slowly and
crossing the legs.
• Increasing sodium intake up to 10g (185mmol) per day and fluid intake
to 2.0–2.5L per day (caution in elderly patients with heart failure).
• Raising the head of the bed by 10–20°, as this stimulates the renin–
angiotensin–aldosterone system and results in a decrease in the
nocturnal diuresis.
• Drinking approximately 500mL of water, which stimulates a significant
pressor response and improves symptoms of postural hypotension.
• Using custom-fitted elastic stockings extending to the waist.
• Pharmacological treatment with fludrocortisone, starting at
100 micrograms per day, whilst carefully monitoring for supine
hypertension, ankle oedema, and hypokalaemia. Potassium
supplementation may be required when higher doses are used, and it
is important to monitor urea and electrolytes.
• In severe cases, the following drugs: alpha-1 adrenal receptor agonist
midodrine (2.5–10.0mg tds), sympathomimetic ephedrine (25mg tds),
and occasionally octreotide and erythropoietin (25–75U per kg three
times a week until a haematocrit level approaching normal is achieved),
may be tried.
Erectile dysfunction
Libido is not normally affected and pain is also unusual, so look for hypog-
onadism and Peyronie’s if they are present. Autonomic neuropathy is the
likely cause, but many drugs, especially thiazides and B-blockers, can also
cause it, as can alcohol, tobacco, cannabis, and stress. These should be
assessed by direct questioning. Examination should include:
• Genitalia and s sexual characteristics.
• Peripheral pulses—as vascular insufficiency may play a part.
• Lower limb reflexes and vibration thresholds—to confirm that
neuropathy is present.
Biochemical screening should at least include:
• Prolactin.
• Testosterone.
• Gonadotrophins (LH/FSH).
Exacerbating factors, such as alcohol and antihypertensive drugs, should
be modified. The main therapies are:
• Oral therapies include: sildenafil (start at 25–50mg, i to 100mg if
needed, and taken 1h prior to sexual intercourse), vardenafil (start
at 10mg, i to 20mg if needed, and taken 25–60min prior to sexual
intercourse), tadalafil (start at 10mg, i to 20mg if needed, and taken
30min to 12h prior to sexual intercourse).
• Intraurethral alprostadil (start at 125 micrograms, i to 250 or 500
micrograms if needed).
• Intracavernosal alprostadil (trial dose is 2.5 micrograms; treatment is
5–40 micrograms).
• Vacuum devices.
802 CHAPTER 13 Diabetes
Macrovascular disease
People with diabetes have a significantly greater risk of macrovascu-
lar complications (coronary heart disease, cerebrovascular disease, and
peripheral vascular disease) than the non-diabetic population. Around
three-quarters of people with diabetes will die as a result of macrovascular
disease, and a diagnosis of type 2 diabetes equates to a cardiovascular risk
equivalent of ageing 10–15 years.
Epidemiology
The exact prevalence and incidence of macrovascular disease and its out-
comes will vary, depending on the age, gender, and ethnic mix of the
patients being assessed. The previous belief that cardiovascular disease is
less common in type 1 diabetes has been proven wrong, and, in general,
vascular complications account for around three-quarters of deaths in
both type 1 and type 2 diabetes. Although the atheroma seen is histologi-
cally the same as in a non-diabetic population, it tends to be more diffuse
and progresses more rapidly. It also occurs at an earlier age, and women
with diabetes lose the cardiovascular protection seen in the non-diabetic
population.
• Overall, peripheral vascular disease occurs in up to 10% of patients,
and they have up to 15-fold greater risk of needing a non-traumatic
amputation than the non-diabetic population.
• Thromboembolic cerebrovascular events increase in individuals with
diabetes, compared to the non-diabetic population.
• The risk of having a myocardial infarction is also increased 2–4 times in
individuals with diabetes.
Secondary prevention
• Stop smoking.
• Strict control of blood pressure.
• Lipid-lowering drugs.
• Early control of blood glucose.
• Aspirin: this is no longer recommended for primary cardiovascular
protection in subjects with diabetes.
Pathogenesis
The risk factors for atherosclerosis, such as smoking and family history, still
apply in a diabetic population. Some factors, however, are more common
in those with diabetes and may also confer a greater risk to the diabetic
population. These include:
• Glycaemic control. Short-term studies, such as ACCORD, VADT, and
ADVANCE, investigating the effects of tight glycaemic control on the
development of macrovascular disease have been disappointing. These
showed that optimizing glucose control has no value in preventing
vascular ischaemic events. However, longer-term studies, such as
DCCT-EDIC in type 1 diabetes and UKPDS in type 2 diabetes, have
clearly demonstrated that early glycaemic control is key for the
prevention of cardiovascular disease later in life. However, strict
MACROVASCULAR DISEASE 811
Management
Patients with diabetes are given dietary advice to help control blood glu-
cose, which also contributes to improving lipid profile. In a patient who is
already following a good ‘diabetic diet’, however, there is often not much
room for improvement.
Other standard advice should also be given:
• Stop smoking—reduces risk of death by about 50% over a 15-year
period.
• Reduce weight if overweight/obese.
• Increase physical activity.
Lipid-lowering therapy
The use of general cardiovascular risk stratification tables is not recom-
mended for individuals with diabetes. NICE guidelines recommend for
those with type 1 diabetes, statin treatment is used in those with raised
albumin excretion rate or ≥2 features of metabolic syndrome or other
risk factors (e.g. age >35 years or high-risk ethnic group). For those with
type 2 diabetes, statin treatment is recommended in all those >40 years
old, unless their CV risk is low (<20% over 10 years, using the UKPDS risk
engine), and in those under 40 years with multiple CV risk factors.
816 CHAPTER 13 Diabetes
Hypertension
Epidemiology
Hypertension is twice as common in the diabetic population as in the
non-diabetic population, and standard ethnic differences in the prevalence
of hypertension still hold true. It is known that hypertension worsens
the severity and increases the risk of developing both microvascular and
macrovascular disease. Using a cut-off of >160/90mmHg, hypertension
occurs in:
• 10–30% of patients with type 1 diabetes.
• 20–30% of microalbuminuric type 1 patients.
• 80–90% of macroalbuminuric type 1 patients.
• 30–50% of Caucasians with type 2 diabetes.
Using the UKPDS suggested target of 140/80, hypertension is even more
common.
Pathogenesis
• Type 1 patients. Hypertension is strongly associated with diabetic
nephropathy and microalbuminuria and occurs at an earlier stage than
that seen in many other causes of renal disease.
• Type 2 patients. Hypertension is associated with insulin resistance and
hyperinsulinaemia. Hyperinsulinaemia can directly cause hypertension
by increased sympathetic nervous system activity, enhanced proximal
tubule sodium reabsorption, and stimulation of vascular smooth
muscle cell proliferation.
HYPERTENSION 819
820 CHAPTER 13 Diabetes
Management of hypertension
Treatment aim
NICE guidelines recommend all patients with diabetes should have a blood
pressure <140/80mmHg or 130/80 in the presence of diabetic complications.
The hypertension study in the UKPDS highlights the benefits for type 2
patients of such a treatment level on mortality, diabetes-related endpoints,
and microvascular endpoints. In this study, a 10/5mmHg difference in BP
was associated with a 34% risk reduction in macrovascular endpoints, a
37% risk reduction in microvascular endpoints, and a 44% risk reduction
in stroke. The Hypertension Optimal Treatment (HOT) study supports
these targets.
Predisposing conditions
Other conditions causing both hypertension and hyperglycaemia should be
considered, e.g. Cushing’s syndrome, acromegaly, and phaeochromocytoma.
End-organ damage
Assess evidence of end-organ damage (eyes, heart, kidneys, and peripheral
vascular tree, in particular).
Assessment of cardiac risk factors
Treat associated risk factors for coronary heart disease.
Treatment
General
Modify other risk factors, such as glycaemic control, smoking, and dyslipi-
daemia. Then consider:
• Weight reduction if obese.
• Reduced salt intake (<6g/day).
• Reduced alcohol intake (<21 units/week in ♂, <14 in ♀).
• Exercise (20–40min of moderate exertion 3–5 times/week).
Pharmacological
Most agents currently available will drop systolic BP by no more than
20mmHg at most. In the UKPDS BP study, one-third of those achieving
the current BP targets required three or more drugs. Recently, the NICE
guidelines for BP treatment were updated and now advocate an ‘A/CD’
approach. That is starting with ‘A’, an ACEI (or an angiotensin II recep-
tor blocker/antagonist if the ACEI is not tolerated), and then adding in
either a ‘C’/calcium channel blocker or a ‘D’/thiazide-type diuretic, with an
A-blocker, a B-blocker, or further diuretic therapy then added if this fails
to reduce BP adequately.
• In the presence of microalbuminuria or frank proteinuria, an ACEI is
first-line, or an angiotensin II receptor antagonist if not tolerated.
• In Afro-Caribbean patients, ACEI and B-blockers are less effective than
calcium channel blockers and diuretics. A diuretic may be needed to
improve the efficacy of the ACEI in these patients.
• Several agents, such as high-dose thiazides and B-blockers, can worsen
diabetic control and exacerbate dyslipidaemia, so tailor the drugs
MANAGEMENT OF HYPERTENSION 821
Lipids and
hyperlipidaemia
Primary hyperlipidaemias
Background
A primary hyperlipidaemia is defined by a knowledge of a monogenic or
polygenic cause. Although the clinical features of p hyperlipidaemia are
driven by a specific defect, the phenotype is also susceptible to environ-
mental pressure. Some of the primary hyperlipidaemias are very aggressive
and need strict clinical attention. At present, the family history and the
phenotypic findings in other family members are used as a surrogate for
a genetic diagnosis. DNA-based diagnosis is established for type III hyper-
lipidaemia and is being implemented for familial hypercholesterolaemia.
The latter can be useful not just for initial diagnosis, but also for family
screening.
Severe isolated hypertriglyceridaemias are classically recessive, and
genetic diagnosis is of little help or unavailable in clinical routine.
POLYGENIC HYPERCHOLESTEROLAEMIA 829
Polygenic hypercholesterolaemia
This is the most common cause of isolated hypercholesterolaemia. LDL
clearance appears to be reduced by a variety of mechanisms, and the E4
allele of apo E is a common association. Patients do not have the charac-
teristic xanthelasmata or extensor tendon deposits (xanthomata) seen in
familial hyperlipidaemia, and occasionally it can be difficult to distinguish
from familial hypercholesterolaemia. If young offspring of cases with poly-
genic hypercholesterolaemia are tested, it would be unsurprising if they
have normal cholesterol concentrations, which is in contrast to children
of those with familial hypercholesterolaemia.
830 CHAPTER 14 Lipids and hyperlipidaemia
Familial combined
hyperlipidaemia (FCHL)
• FCHL is a high-risk syndrome for CHD.
• Population frequency has been estimated to 1/200.
• The aetiology is not yet known. Despite its seemingly dominant
appearance, the risk is clearly conveyed by a number of different
unknown genes.
• It is the most common type of inherited dyslipidaemia, estimated to
cause 10% of cases of premature CHD.
• It has no unique clinical manifestations, and the diagnosis is based on
raised lipids (>95th centile for age) and a family history of premature
CHD in first-degree relatives. In contrast to FH, tendon xanthomata
should not be present.
• The lipid phenotype can vary within individuals and family members
such that combined hyperlipidaemia, isolated hypercholesterolaemia,
and isolated hypertriglyceridaemia can be seen within the same
individual at different times and in different family members.
• Very likely to have raised Apo B concentrations.
• Should be treated aggressively and very often needs the combination
of several different pharmacological agents.
Familial hypertriglyceridaemia
• Is defined as a subentity of FCHL, but clearly less common, perhaps
with a frequency of 1/1,000, often as an autosomal dominant trait with
elevated VLDL levels, and is frequently accompanied by a moderate
hypercholesterolaemia. Triglyceride concentrations are rarely higher
than 10mmol/L.
• Suspect this condition where there is a subtle hypertriglyceridaemia
without any obvious causes (obesity, type 2 diabetes, etc.), together
with a positive CHD or stroke family history.
• The condition can sometimes be surprisingly responsive to statins.
FAMILIAL DYSBETALIPOPROTEINAEMIA 833
Familial dysbetalipoproteinaemia
• Also known as type III hyperlipidaemia or broad beta disease.
• It is associated with early-onset CHD.
• It is an uncommon disorder affecting 1/10, or less, of people with the
Apo E2/E2 genetic background (which is 1/100 in most populations).
• The condition leads to specific elevation of IDL and chylomicron
remnants which are both cholesterol- and triglyceride-rich. Typically,
the rise of cholesterol and triglycerides is equimolar, and very high
concentrations can be seen (15mmol/L of TG and 15mmol/L of
cholesterol).
• Diagnosis is confirmed by genetic testing.
• A characteristic clinical feature is the presence of palmar striae
xanthoma; tubero-eruptive xanthomata, found over the tuberosities of
the elbows and knees, may also be present.
• Due to the recessive nature of the apo E2 genotype and the required
additional environmental pressure, family history is rarely revealing.
• Apo E2/E2 genetic background is not enough to precipitate the
syndrome; an additional factor is needed. This is typically obesity, type
2 diabetes, hypothyroidism, B-blocker, or thiazide diuretic medication.
The oral contraceptive pill can also elicit type III hyperlipidaemia.
834 CHAPTER 14 Lipids and hyperlipidaemia
Elevation of Lp(a)
• Lp(a) is an LDL-like lipoprotein particle, but it is not cleared via the
LDL receptors. Less than 5% of the population have raised Lp(a),
and the distribution is very skewed. Synthesis appears to be highly
genetically driven. Recent evidence suggests this is a highly atherogenic
lipoprotein particle. Raised Lp(a) should be suspected in someone
with early presentation of CHD, stroke, or peripheral vascular disease,
with moderately elevated cholesterol that is seemingly resistant to
statin therapy. At present, the only available pharmacological means by
which lowering of Lp(a) can be achieved is with niacin. However, there
is no formal evidence to suggest that niacin reduces cardiovascular
events in people with raised Lp(a).
RARE FAMILIAL MIXED DYSLIPIDAEMIAS 835
Secondary hyperlipidaemias
Background
Secondary hyperlipidaemias are common, and the predominant precipitat-
ing factors are obesity and type 2 diabetes They may consist of isolated
elevations of cholesterol or triglycerides or be combined. Treatment is
based upon managing the primary disorder disease before making a fur-
ther decision on the raised lipids. Not infrequently, more than one cause
is apparent in secondary hyperlipidaemias.
Causes
(Summarized in Table 14.1.)
• Obesity (b see Consequences of obesity, p. 857).
• Diabetes mellitus (b see Lipid abnormalities found in patients with
diabetes, p. 814).
• Fatty liver disease. Almost invariably associated with elevated TG.
• Diet. Excessive consumption of saturated fats, carbohydrate, sugary
drinks, and alcohol. Raised triglycerides by alcohol are normally
only seen after excessive consumption. Occasionally, the unusual
combination of elevated HDL cholesterol with raised triglycerides
is seen.
• Hypothyroidism. Estimated to occur in 4% of those with
hyperlipidaemia, and compensated (subclinical) hypothyroidism in a
further 10%. Usually resulting in hypercholesterolaemia with a TC of
7–12mmol/L. It also worsens primary hypercholesterolaemias due to d
synthesis of hepatic LDL receptors.
• Chronic renal disease.d creatinine clearance is accompanied by
hypertriglyceridaemia and d HDL cholesterol. Proteinuria, in the
nephrotic syndrome, is associated with hypercholesterolaemia.
It also raises Lp(a). After kidney transplantation, ciclosporin may
increase LDL.
• Liver disease. Especially with cholestasis, resulting in abnormal LDL
cholesterol. Primary biliary cirrhosis often results in TC >12mmol/L,
which is close to treatment resistance. The accumulating cholesterol
is contained in a fraction called LpX, absent in healthy humans. These
are large aggregates of phospholipids and free cholesterol and do not
resemble conventional lipoproteins. However, severe hepatocellular
damage may also lower LDL cholesterol by d production of its
component parts and the enzymes which metabolize it.
• Cushing’s syndrome. Glucocorticoids increase VLDL production,
thus hypertriglyceridaemia. The associated weight gain and glucose
intolerance can make this effect more pronounced.
• Lipodystrophies. A very rare group of disorders, hallmark of
which is regional, partial, or generalized fat loss, associated with
hyperlipidaemia, especially unusually raised triglycerides. Also
associated with glucose intolerance. See b Genetic causes of severe
insulin resistance, p. 693.
SECONDARY HYPERLIPIDAEMIAS 837
Management of dyslipidaemia
Background
Hyperlipidaemias are common, and a decision to use pharmacological
means to lower hyperlipidaemias should depend on estimated cardiovas-
cular risk. For this purpose, useful risk scoring systems have been devel-
oped, but it is important to use these wisely. Importantly, they should
not be used for certain primary and familial conditions with very high
cardiovascular risk.
It can be a challenge to the physician to describe benefits of the medica-
tion or the necessary change in lifestyle to the patient, as treatment rarely
comes with an obvious ‘reward’ and the true effect may only be described
as a certain CHD risk reduction noticeable a decade or more ahead.
Overall, the use of statins has an extremely good evidence base, whereas
most alternative or complementary options have less solid evidence.
Primary and secondary prevention
It is universally accepted that lowering hypercholesterolaemia using statins
in secondary prevention reduces future cardiovascular events and overall
mortality. It is estimated that a 10% fall in TC results in a 25% decrease
in CHD risk. Regression and remodelling of atheromatous lesions have
also been clearly demonstrated. The presence of type 2 diabetes mellitus
puts the patient in the category for secondary prevention. Acute coronary
syndrome should be treated aggressively (high-dose atorvastatin 80mg),
which shows early benefit with statin treatment. In post-MI patients,
omega-3 fatty acids are recommended, as it reduces the incidence of sud-
den death due to arrhythmias.
A decision to reduce cholesterol primary prevention is much depend-
ent on the overall cardiovascular risk (b see p. 826). The reduction
in absolute risk is marginal in patients with few other risk factors and
should be evaluated critically. An estimated risk of CHD >2% per annum
has unquestionable support for initiation of pharmacological treatment.
However, even young patients with primary and familial conditions should
be treated aggressively without applying risk scoring systems.
Specific interventions
Dietary advice
Current recommendations are that fats should constitute <30% of energy
consumed and saturated fats must be <30% of the total fat content. To
achieve this, vegetable and marine mono- and polyunsaturated fats should
be high in the diet, whereas animal and dairy fat should be low. Total
dietary cholesterol should not exceed 300mg per day. Foods advocated
include fresh fruit and vegetables, which are also important sources of
antioxidants. A high fibre content is cholesterol-lowering. Four months
should be given to see if dietary manipulation will work in patients with
low-to-moderate risk. It is unusual to see more than a 15% fall in choles-
terol from dietary measures, but certain individuals can respond better.
Alcohol should be <14 units/week for a ♀ and <21 units/week for a ♂.
MANAGEMENT OF DYSLIPIDAEMIA 839
Plant sterols and stanols, 2–3g daily, can reduce blood cholesterol by
10–15%. They are available commercially in enriched margarine spreads,
yoghurt, and milky drinks.
Weight control
All overweight patients must be encouraged to lose weight. Weight
reduction is closely attuned to dietary advice and physical exercise. Most
weight is lost in the first 4 months of a regimen. A 10kg weight loss in
an obese subject can reduce LDL cholesterol by 7% and increases HDL
cholesterol by 13%. The effect on raised triglycerides can be much greater
and clinically very useful.
Physical activity
Physical activity, especially aerobic exercise, is recommended. This should
be realistic and adapted to the individual’s capacity. Although exercise
levels in the range of 70% of VO2max for 30–40min at least 3–5 times per
week is extremely useful, it is often unrealistic, and very good effects are
seen by walking, etc. Acute exercise will transiently change lipoprotein
levels and increase lipoprotein lipase activity. These effects become more
permanent with regular training. Triglyceride levels fall, HDL cholesterol
levels rise, especially the HDL2 subfraction with more vigorous exercise,
and the LDL cholesterol is of the less dense variety which is not so ath-
erogenic. The changes are dose-dependent with i exercise, and a 20%
alteration in each variable is achievable after 6 weeks. Particularly good
effects are seen in type 2 diabetes mellitus.
Modification of other risk factors
Other risk factors, such as hypertension, smoking, and diabetes mellitus,
must be addressed.
840 CHAPTER 14 Lipids and hyperlipidaemia
Drug therapy
Numerous agents can be used for both p and s prevention in patients in
whom non-pharmacological approaches have either been unsuccessful or
deemed insufficent.
HMG CoA reductase inhibitors (statins)
Table 14.2 shows the comparative potency of common statins.
Indications
Raised LDL, heterozygous + homozygous FH, mixed hyperlipidaemia. Not
all these medications are licensed for use in children and should be used
with caution in ♀ of childbearing age because of potential teratogenicity.
The medication should be stopped for at least 3 months before pregnancy
is planned. Women of childbearing age must ensure effective contracep-
tion when on these drugs.
Mechanism of action
Inhibition of 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) which
is the rate-limiting enzyme in cholesterol synthesis. Its activation increases
hepatocyte LDL receptor numbers which leads to increased clearance of
LDL and VLDL remnants. See Table 14.2 for comparative lipid-lowering
profile and Table 14.3 for dosage.
Side effects
Statins are usually very well tolerated. The most common side effect is
muscle ache which, in extremely rare cases, also develops into myositis.
Hepatotoxicity is not common but does exist. LFTs and creatine kinase
should be measured before they are prescribed and used with caution in
those with an excessive alcohol intake. It is recommended that the medi-
cations should be discontinued if the liver enzymes aspartate transami-
nase (AST) and/or alanine transaminase (ALT) show more than a 2–3-fold
elevation above the upper limit of normal. LFTs should, therefore, be
checked within 3–4 months of treatment and, at most, annually in the long
term if the patient is without symptoms. Myositis is rare, but the frequency
increases with concomitant medications, e.g. ciclosporin, fibrates, or when
renal impairment or untreated hypothyroidism are present. Clinically,
there is a picture of swollen tender muscles and creatine kinase being
>10× the upper limit of normal. Rhabdomyolysis is even rarer.
Interactions
Several statins are metabolized through CYP3A4. Grapefruit juice should
be avoided. Statins can interact with ciclosporin. There is an interaction
with warfarin for atorvastatin, simvastatin, and rosuvastatin, but it is often
not clinically significant and would only lead to small adjustments of the
warfarin dose. Erythromycin interacts with all statins. Digoxin interacts
with atorvastatin and simvastatin. Rifampicin interacts with fluvastatin and
pravastatin. Atorvastatin and rosuvastatin may also interact with the oral
contraceptive pill, antacids, and some antifungals.
DRUG THERAPY 841
Interactions
Use with caution in combination with statins. Can enhance the effects of
warfarin and antidiabetic agents and is contraindicated in those on orlistat.
Anion exchange resins (bile acid sequestrants)
Indications
High LDL, i.e. hypercholesterolaemia. Largely superseded by statins, these
are now best used as adjuncts when LDL has not fallen enough with a
statin alone. They are also the only drug licensed for use during pregnancy.
Mechanism of action
Bind to bile acids in the gut, so reducing their enterohepatic circulation
and i bile acid excretion. This increases hepatocyte cholesterol require-
ments which increases LDL receptor production, so reducing circulating
LDL levels. Under optimum conditions, LDL cholesterol can be reduced
by 20–30%; triglycerides rise by 10–17%, and HDL increases by 3–5%.
Side effects
These agents remain in the gut, so constipation, bloating, nausea, and
abdominal discomfort are not uncommon. Constipation, found in 35–40%
of those on these agents, can be helped with bulking laxatives. Less often,
a bleeding tendency due to vitamin K malabsorption can be seen. These
agents can also exacerbate hypertriglyceridaemia, but HDL cholesterol
is often i slightly. To reduce the side effects, start with low doses and
build up gradually over the next 3–4 weeks while maintaining a good fluid
intake.
Interactions
These agents can reduce the absorption of warfarin, digoxin, β-blockers,
pravastatin, fluvastatin, and hydrochlorothiazide. Many other agents, such as
simvastatin, have not been checked, so to avoid any potential interaction,
advise patients to take all other drugs 1–3h before or 4–6h after the resin.
Nicotinic acid and acipimox
Indication
High LDL, VLDL, IDL, or triglycerides. Nicotinic acid is the most effective
medication for i HDL cholesterol. In practice, however, their use is lim-
ited by the side effect profile, especially flushing.
Mechanism of action
Work by inhibiting lipolysis in adipocytes, so altering fatty acid flux and
reducing VLDL synthesis and HDL clearance, or by inhibiting VLDL tri-
glyceride synthesis. Plasma triglycerides fall by 20–50%, and LDL by 5–25%
while HDL levels rise (by 10–50%).
Side effects
Common, with 30% unable to tolerate these agents. Vasodilatation, giving
cutaneous/facial flushing, occurs in most patients but tends to improve
after 2–3 weeks of therapy. Pruritus and dry, burning sensation in the skin
can be seen. High doses of aspirin were previously given to reduce this
effect, but this is now rarely practised due to the side effects of high-dose
DRUG THERAPY 843
aspirin. Patients should also be instructed to avoid hot drinks and spicy
food. Gastritis is also a common side effect, whereas liver side effects
are rare. Raises uric acid almost invariably, and exacerbation of gout can
be seen. Rarely, precipitation of acanthosis nigricans and retinal oedema
are also recognized side effects. Nicotinic acid can adversely affect glucose
control in prediabetic states and in diabetes mellitus. Acipimox, a nicotinic
acid analogue, seems to be less problematic but is also less potent. In 2013
following the results of the HPS2-THRIVE study, the European Medicines
Agency suspended licences for nicotinic acid preparations on the grounds
that they were ineffective and associated with adverse effects. Acipimox
remains available.
Omega-3 fatty acids (Omacor®, Maxepa®)
Indications
Hypertriglyceridaemia.
Mechanism of action
Inhibit the secretion of VLDL due to i intracellular apo B-100 destruc-
tion. Normal patients see both a fall in VLDL and LDL, but LDL may rise
in the hypertriglyceridaemic individual. Should be seen as add-on therapy
to statins. Lower doses (1g per day) have demonstrated positive effects
on cardiovascular prevention, but the mechanism of action is likely to be
through an antiplatelet effect.
Side effects
As high doses are needed, this is a high calorie load and may increase
obesity. More commonly, gives nausea and belching. Omacor® produces
fewer unwanted effects and is less calorific, as it is used in a smaller dose.
Interactions
None significant.
Cholesterol absorption blocker
Ezetimibe is the only available compound. Should be used as second-line
treatment after a statin.
Indications
LDL cholesterol-lowering in patients who are intolerant of statins or in
combination with a statin in those that are not adequately controlled with
a statin alone. Also in the very rare condition of sitosterolaemia where
there is an i absorption of plant sterols.
Mechanism of action
Dietary and biliary cholesterol absorption is selectively inhibited.
Ezetimibe (10mg) monotherapy produces an 18% fall in LDL cholesterol
and an increase in HDL cholesterol of 1–3%, and triglycerides are not
affected. In combination therapy with a statin, ezetimibe reduces levels by
an additional 22% to that obtained by statin alone.
Interactions
None significant.
844 CHAPTER 14 Lipids and hyperlipidaemia
Further reading
Bhatnagar D, Soran H, Durrington PN (2008). Hypercholesterolaemia and its management. BMJ
337, a993.
Chapman MJ, Ginsberg HN, Amarenco P (2011). European Atherosclerosis Society Consensus
Panel. Triglyceride-rich lipoproteins and high-density lipoprotein cholesterol in patients at high
risk of cardioivascular disease: evidence and guidance for management. Eur Heart J 32, 1345–61.
Graham I, Atar D, Borch-Johnsen K, et al. (2007). European guidelines on cardiovascular disease
prevention in clinical practice: full text. Fourth Joint Task Force of the European Society of
Cardiology and other societies on cardiovascular disease prevention in clinical practice (consti-
tuted by representatives of nine societies and by invited experts). Eur J Cardiovasc Prev Rehabil
14 Suppl 2, S1–113.
Watts GF, Karpe F (2011). Triglycerides and atherogenic dyslipidaemia: extending treatment
beyond statins in the high-risk cardiovascular patient. Heart 97, 350–6.
Hippisley-Cox J, Coupland C, Vinogradova Y, et al. (2008). Predicting cardiovascular risk in England
and Wales: prospective derivation and validation of QRISK2. BMJ 336, 1475–82.
The National Institute for Clinical Excellence guidelines on lipid modification. Available at: M
http://guidance.nice.org.uk/CG67.
The National Institute for Clinical Excellence guidelines: identification and management of familial
hypercholesterolaemia. Available at: M http://www.nice.org.uk/CG71.
The online access to metabolic and molecular basis of inherited disease. Available at: M http://
www.ommbid.com/OMMBID/the online metabolic and molecular bases of inherited disease/b/
parttoc/part 12.
Chapter 15 847
Obesity
Definition of obesity
Obesity is defined as an excess of body fat sufficient to adversely affect
health. Body mass index (BMI) and waist circumference, as a measure of
fat distribution, are the most commonly used measures, but a clinical stag-
ing system is increasingly used to determine risk and management (see
Box 15.1). BMI is an imprecise measure of adiposity and does not account
for fat distribution, which may better determine metabolic and cardiovas-
cular risk at lower BMI.
Lower cut-off values for BMI and waist circumference are applicable to
non-Caucasian ethnic groups: cut-off points for increased risk varies from
22kg/m2 to 25kg/m2 in different Asian populations, and for high risk from
26kg/m2 to 31kg/m2.
Central obesity may reflect increased visceral (intra-abdominal fat)
stores and/or ‘ectopic’ fat (fat stored in liver, muscle, pancreas, and epicar-
dium) more directly linked to pathophysiology, such as insulin resistance.
Assess overall clinical status, using BMI, waist circumference, and stage
(see Box 15.1).
Obesity is associated with increased secretion of adipose tissue prod-
ucts, including hormones, cytokines (adipocytokines), and growth factors
such that it is now regarded as a disease of chronic low-grade systemic
inflammation. Many of its adverse sequelae relate to this pathology.
DEFINITION OF OBESITY 849
Epidemiology of obesity
Overweight and obesity are rapidly increasing globally; the highest rates
of obesity (>30%) are seen in the USA, Mexico, and the Middle East. One
billion adults are currently overweight (BMI 25–29.9kg/m2), and a further
475 million are obese. When Asian-specific cut-off points for the definition
of obesity are taken into account, the number of adults considered obese
globally is over 600 million.
In the UK, prevalence has doubled in 25 years: 26.1% of adults (aged
16 years and over) were obese in 2010, and 55% of the population is either
overweight or obese.
Global estimates of childhood obesity suggest 200 million school-aged
children are either overweight or obese, of whom 40 million are obese.
In the UK, 10.1% of boys and 8.8% of girls aged 4–5 years and 20.6% of
boys and 17.4% of girls aged 10–11 years are obese
Prevalence of obesity varies with age (peak prevalence at 50–70 years),
socio-economic class (15% women and 21% men in social class I; 34% and
30% in social class V) and social deprivation in women (22% least to 31%
most deprived).
EPIDEMIOLOGY OF OBESITY 851
852 CHAPTER 15 Obesity
Aetiology of obesity
Overweight and obesity results from a complex interaction between envi-
ronmental pressures and risks and genetic susceptibility. Heritability of
obesity is about 60%, but the rapid increase in obesity prevalence over
the past 30 years argues in favour of predominantly environmental drivers.
However, there is increasing interest in the possibility of epigenetic influ-
ences on obesity related to maternal obesity, diet, gestational diabetes,
and even possibly environmental pollutants, such as polyfluorinated com-
pounds and polycyclic aromatic hydrocarbons.
Genetic factors
Monogenic obesity
Mutations in genes (usually related to appetite control within the hypo-
thalamus) are associated with obesity of early childhood onset, usually
with hyperphagia. However, only about 5% of all severe childhood and
2% of adult obesity are associated with identified genetic causes. Of these,
mutations in the melanocortin 4 receptor (MC4R) are the most frequent
and are associated with increased linear growth, fat and lean mass, hyper-
phagia (moderate) and severe hyperinsulinaemia, but normal puberty and
fertility.
An increasing number of genes associated with the development of obe-
sity have been identified through genome-wide association studies. The
FTO ‘fat mass and obesity associated’ gene was originally linked to type 2
diabetes; the association was actually found to be due to the higher BMI of
diabetic cases in comparison to non-diabetic controls; common variants in
the first intron result in a +0.4kg/m2 elevation in BMI per risk allele. Similar
to the situation with type 2 diabetes, possession of ‘risk’ single nucleotide
polymorphisms (SNPs) only accounts for a small increase in susceptibility
to obesity or adult BMI.
Environmental factors
The drivers of obesity can be considered under two main head-
ings: increased energy (food) intake or decreased energy expenditure due
to physical inactivity, as the major determinants of obesity in genetically
susceptible individuals. Societal changes, e.g. increased availability of high
caloric density foods and sedentary lifestyle, have been one of the main
causes of changes in this balance.
Secondary causes
These are uncommon because, even when linked to weight gain, the dis-
ease usually manifests itself from its particular pathophysiology.
• Hypothyroidism: an important cause to exclude in children but rarely
presents simply with weight gain in adults.
• Cushing’s disease/syndrome: rare, but an important cause to exclude in
obese patients presenting with ‘overlap’ signs and symptoms which can
include striae, depression, and hypertension.
AETIOLOGY OF OBESITY 853
Pathophysiology of obesity
Energy balance and body weight are regulated, but the main drive of this
allostatic physiology is towards energy acquisition (and thus fat deposition)
and defence against weight loss. Although physiology can be ‘overridden’
by cognitive and behavioural control (e.g. diet, exercise) long-term, these
mechanisms (within our obesogenic environment) usually prove insuffi-
cient in the long-term either to protect against or reverse weight gain.
Long-term signals associated with body fat stores are provided by leptin
and insulin. In human obesity, leptin levels are high, rather than low, cor-
relating with fat mass, suggesting either that leptin ‘resistance’ is present
or that leptin is a starvation, rather than obesity, signal (see Box 15.2 and
Table 15.1).
Gut peptide hormones released after food intake provide acute signals
of hunger, satiety, and fullness (see Box 15.3). Although originally thought
to be short-term signals, the importance of the gut–brain axis as a regu-
lator of body weight in humans has become increasingly apparent from
the effects of bariatric surgery. Ghrelin, a hunger hormone, rises before,
and probably is involved with, initiation of food intake. Satiety hormones
released after food include glucagon-like peptide-1 (GLP-1), an incretin
hormone secreted by ileal L-cells in the distal intestine that stimulates
insulin secretion, and peptide YY secreted from the ileum and colon.
Oxyntomodulin (also derived from preproglucagon) reduces food intake
and increases energy expenditure after systemic administration.
Weight is gained or lost usually in the proportion of 70% fat and 30%
lean tissue, implying that approximately 30MJ (7,000kcal) surplus or deficit
is needed to gain or lose 1 kg in body mass. During the first days of a very
low energy or ketogenic diet, liver glycogen may be the primary source
of stored energy to meet metabolic needs; since it provides about 8MJ/kg,
initial weight loss is more rapid than with less severe energy restriction.
PATHOPHYSIOLOGY OF OBESITY 855
Consequences of obesity
Most metabolic, physiological, and organ systems are affected by obe-
sity and can be considered under the ‘4 M’ headings: mental, metabolic,
mechanical, and monetary (see Box 15.4).
• Mortality rates rise steadily at BMI >5kg/m2. Obesity and physical
inactivity have both independent and dependent effects on all-cause
mortality.
• Loss of life expectancy: BMI >35kg/m2—5–7 years at age 45.
• Type 2 diabetes: elevation in BMI, the dominant risk factor for
development of diabetes. Relative risk (RR) in overweight men 2.4,
women 12.4; at BMI 30kg/m2 >10; increased to 50–90-fold at BMI >35.
• Hypertension: RR for overweight men 1.8, women 2.4.
• Dyslipidaemia: moderate relationship with total cholesterol, closer
relationship with triglycerides, HDL cholesterol.
• Stroke: RR 1.2 for overweight and 1.5 for obese men and women.
• Asthma: obese 2 ×, overweight 1.4 × more likely to develop asthma.
Additional tests
• Liver ultrasound if abnormal liver function to confirm NAFLD.
• Echocardiogram if heart failure suspected.
• Pharmacological stress testing if ischaemic heart disease suspected
(subject may be unable to undertake exercise test).
• Endoscopy if anaemia—gastroscopy if reflux disease, colonoscopy if
colon cancer suspected.
• Gynaecological referral if post-menopausal bleeding (high incidence of
endometrial cancer in obese women).
• Dexamethasone suppression (overnight or low-dose) if Cushing’s
disease suspected.
• Transferrin, then genetic testing for haemochromatosis in patients with
type 2 diabetes and abnormal liver function.
Advise on risks of obesity, benefits of weight loss and treatment options,
and the need for a long-term strategy. Treatment options include:
• Stress management and self-assertiveness training.
• Dietary intervention.
• Physical activity.
• Psychological counselling.
• Anti-obesity medication.
• Bariatric surgery.
Agree on:
• Weight loss goals.
• Behavioural goals and health outcomes.
• Management plan.
Assist in addressing drivers and barriers to weight management; offer
referral to appropriate provider (primary care for diet and lifestyle pro-
grammes, psychology services if binge eating disorder or severe depres-
sion, secondary care if obesity severe or complex).
General principles
Dietary energy restriction produces greater weight loss than exercise.
Increased physical activity and exercise predicts weight loss maintenance.
Patients’ expectations frequently exceed realistic goals or need.
A weight loss of 5–10% of the initial body weight reduces many
of the health risks associated with obesity and reduces cancer and
diabetes-related mortality; a 5kg loss in adults with prediabetes reduces
progression to diabetes by 60%, maintained even if weight regain occurs. In
patients with a BMI >35kg/m2, greater weight loss (>10kg or 15%) is likely
to be needed to produce a sustained improvement in comorbid diseases;
10–15% may be needed.
Diet, physical activity, and behavioural therapy
• Dietary advice should aim to reduce energy intake to produce a 2.5MJ
(600kcal)/day deficit (calculated from standard equations of resting
energy expenditure and assuming a physical activity level (PAL) of
1.3). Both low carbohydrate (<30g/day) and low fat (<30% total daily
energy) produce equivalent weight loss (4–5kg) at 1 year. High-protein
diets may be more satiating.
860 CHAPTER 15 Obesity
Bariatric surgery
Three types of surgery are commonly performed—all usually done lapa-
roscopically. The most compelling data for the success of bariatric surgery
at producing weight loss and improving the clinical outcomes for obese
patients come from the 20-year follow-up data of the Swedish Obese
Subjects study (a case control study started at a time when surgical tech-
niques were not as advanced as nowadays). The persistent weight loss in
the surgical groups was associated with a much reduced mortality: the
unadjusted overall hazard ratio was 0.76 in the surgery group (P = 0.04),
compared with the control group, and the hazard ratio adjusted for sex,
age, and risk factors was 0.71 (P = 0.01). Other studies have confirmed
that bariatric surgery is associated with reduced all-cause mortality, includ-
ing deaths from CVD, diabetes, and cancer.
In patients with type 2 diabetes, benefit extends beyond weight loss. Up
to 80% of people with type 2 diabetes may experience remission of their
diabetes (normoglycaemia without the need for hypoglycaemic medica-
tion), the exact remission rate being determined by the type of surgery
and the duration of diabetes prior to surgery.
Gastric bypass (see Fig. 15.1)
Patients will lose approximately 50–75% of their excess weight (30–40%
absolute weight loss) within 12–18 months of surgery. The large majority
(>80%) of patients will have a significant improvement or resolution of
their weight-related illnesses, and most patients report dramatic changes
to their quality of life. Routine supplementation of vitamins, minerals, and
vitamin B12 is required.
The gastric bypass works in the following way:
• There is a restriction in food intake, as the stomach is reduced to the
size of a large egg.
• Food bypasses digestive secretions, causing some malabsorption.
• Profound changes are seen in gut hormones that control hunger,
satiety, and glucose metabolism (reduced ghrelin, increased and earlier
release of GLP-1 and PYY).
• Other mechanisms, involving changes in gut flora and bile salt
metabolism that affect appetite and CV risk, are also postulated.
Sleeve gastrectomy (see Fig. 15.2)
Patients will lose approximately 50–75% (30–40% absolute weight loss)
of their excess weight within 12–18 months of surgery. Again, >80% have
a significant improvement in weight-related illnesses and improved QoL.
Routine supplementation of vitamins is required for the first year.
The sleeve gastrectomy works in the following way:
• 75% of the stomach is removed, restricting the volume of food patients
are able to eat in one sitting.
• Gastric emptying is faster which alters gut hormone profiles, resulting
in satiety (reduced ghrelin, increased and earlier release of GLP-1
and PYY).
BARIATRIC SURGERY 863
Gastric Bypass
Sleeve Gastrectomy
Pitfalls in laboratory
endocrinology
Introduction 868
Analytical factors 870
Post-analytical factors 872
868 APPENDIX 1
Introduction
As with all biochemical investigations, their usefulness lies in careful and
appropriate selection, combined with discerning interpretation. Where
neither is possible nor simple, the chemical pathologist, or other members
of the analysing laboratory, would always be happy to help.
The results obtained by the wide variety of analytical techniques can
be altered by many factors, some associated with an underlying pathol-
ogy and others not. A brief consideration of potential confounding factors
seems relevant here.
Analytical factors
Most endocrine assays are immunoassays which rely on binding of an
analyte by a diagnostic antibody. The binding specificity will depend on
the care and attention with which the manufacturer has chosen the rea-
gents. Typical interferences in small molecules are due to slightly differ-
ent molecular forms and typically occur with steroid and digoxin assays.
Peptide hormones are more complex because so many differently glyco-
sylated forms of those peptides exist.
Standards for pituitary hormones are generally derived from purified
pituitary extracts which contain a mixture of peptides. This leads to differ-
ent assays having quite marked biases between each other due to different
binding affinities of the antibodies to the different isoforms. A similar situ-
ation exists with hCG for which many multiple molecular forms co-exist.
Attempts to find international consensus for standards which can be used
in diagnostic systems are being made, particularly for growth hormone and
glycated haemoglobin at the present time.
Analytical performance or assay reproducibility is important in deter-
mining the critical differences between patient samples. A significant dif-
ference between consecutive samples at 95% confidence will require a
difference of 1.96 x the method standard deviation at the appropriate
concentration.
The hook effect occurs when very high analyte concentrations flood
the available antibody in vitro and this leads to artefactually low results.
It is less common nowadays as assays have large dynamic ranges but new
cases continue to be reported, and there are published case reports for all
hormones and tumour markers.
Antibody interference is a widespread problem that affects approxi-
mately 1% of immunoassays. It is insidious and is due to endogenous
antibodies which interfere with analyte binding in vitro. Most importantly,
they cannot be detected by usual quality control mechanisms. There are
a number of laboratory techniques that can be used to clarify whether
such interference is present but it is inherent on the clinician to alert the
laboratory to a potential clinical mismatch.
Some assays can only be classified as problem assays. These include
thyroglobulin and low concentrations of oestradiol (<300pmol/L) and
testosterone (<5nmol/L). The former is due to the high prevalence of
endogenous anti-thyroglobulin antibodies which are particularly prevalent
in patients with thyroid disease. The latter are due to antibody specificity;
however, it is hoped that this will be resolved with the introduction of
mass spectrometry into routine clinical practice.
APPENDIX 1 871
Post-analytical factors
Interpretation of assay results is made in relation to reference ranges
provided by the laboratory. These usually represent the 95th centiles of
a population of healthy individuals. However, the definition of normal-
ity is subjective and reference ranges are affected by such factors as age,
gender, and in some cases by ethnicity. Moreover, for hormones, time of
day, month, and season will be important. For some analytes, it is diffi cult
to obtain appropriate samples to construct ranges such as in children and
circumstances that are difficult to obtain in health e.g. following pharma-
cological stimulation or samples of CSF.
If the central 95th centile reference ranges are used, there is a 5% chance
that a result will be out-of-range due to chance. As the number of tests
taken are i, so will the risk of a chance abnormality. The increase will be
x% (where x = 1 – 0.95a and a is the number of tests performed).
Literature from the USA and European journals may use different
units—beware! SI units use molar or mass (g) and volumes reported in
litres.
Most assays are standardized with international preparations. These
are usually the molecular forms that are most prevalent when basal sam-
ples are taken. However, following stimulation non-standard molecules
are released into the circulation which have different clearance rates and
different binding characteristics to the diagnostic antibodies in vitro. This
can lead to marked differences between methods. For example, following
stimulation by ACTH corticosteroid precursors are released which will
compete with cortisol for binding in the assay; similarly following stimula-
tion of GH release different isoforms of GH are secreted and the most
abundant 20 and 22kDa isoforms clear at different rates leading to varia-
tions in recognition by the diagnostic antibodies at different times during
the test.
Reference intervals
874 APPENDIX 2
Introduction
All values are for serum unless specified otherwise.
96 50 48 45 43 40 38 37 35 33 32 30 29 28 27 26 24 15st 2
94 49 47 44 42 40 38 36 34 33 31 30 28 27 26 25 24 14st 11
92 48 46 43 41 39 37 35 33 32 30 29 28 27 25 24 23 14st 7
90 47 45 42 40 38 36 34 33 31 30 28 27 26 25 24 23 14st 2
88 46 44 41 39 37 35 34 32 30 29 28 27 25 24 23 22 13st 12
86 45 43 40 38 36 34 33 31 30 28 27 26 25 24 23 22 13st 8
84 44 42 39 37 35 34 32 30 29 28 27 25 24 23 22 21 13st 3
82 43 41 38 36 35 33 31 30 28 27 26 25 24 23 22 21 12st 13
80 42 40 38 36 34 32 30 29 28 26 25 24 23 22 21 20 12st 8
78 41 39 37 35 33 31 30 28 27 26 25 24 23 22 21 20 12st 4
76 40 38 36 34 32 30 29 28 26 25 24 23 22 21 20 19 12st
74 39 37 35 33 31 30 28 27 26 24 23 22 21 20 20 19 11st 9
72 38 36 34 32 30 29 27 26 25 24 23 22 21 20 19 18 11st 5
70 37 35 33 31 30 28 27 25 24 23 22 21 20 19 19 18 11st
68 36 34 32 30 29 27 26 25 24 22 21 21 20 19 18 17 10st 10
66 35 33 31 29 28 26 25 24 23 22 21 20 19 18 18 17 10st 6
64 34 32 30 28 27 26 24 23 22 21 20 19 18 18 17 16 10st 1
62 33 31 29 28 26 25 24 22 21 20 20 19 18 17 16 16 9st 11
60 32 30 28 27 25 24 23 22 21 20 19 18 17 17 16 15 9st 6
58 30 29 27 26 24 23 22 21 20 19 18 18 17 16 15 15 9st 2
56 29 28 26 25 24 22 21 20 19 18 18 17 16 16 15 14 8st 11
54 28 27 25 24 23 22 21 20 19 17 17 16 16 15 14 14 8st 7
52 27 26 24 23 22 21 20 19 18 17 16 16 15 14 14 13 8st 3
50 26 25 23 22 21 20 19 18 17 17 16 15 14 14 13 13 7st 12
48 25 24 23 21 20 19 18 17 17 16 15 14 14 13 13 12 7st 8
46 24 23 22 20 19 18 18 17 16 15 15 14 13 13 12 12 7st 3
44 23 22 21 20 19 18 17 16 15 15 14 13 13 12 12 11 6st 13
42 22 21 20 19 18 17 16 15 15 14 13 13 12 12 11 11 6st 9
40 21 20 19 18 17 16 15 15 14 13 13 12 12 11 10 10 6st 4
38 20 19 18 17 16 15 14 14 13 13 12 11 11 11 10 10 6st
36 19 18 17 16 15 14 14 13 12 12 11 11 10 10 9 9 5st 9
4' 61/2 4' 8 4' 91/2 4' 11 5' 1/2 5' 2 5' 4 5' 51/2 5' 7 5' 81/2 5' 10 5' 111/2 6' 1 6' 3 6' 41/2 6' 6
Height (ft/in)
Key:
BMI <20 BMI 20-25 BMI 25-30 BMI 30-35 BMI 35-40 BMI >40
Underweight (19-23 for S Asian) (23-28 for S Asian) (>28 for S Asian) Obese Morbidly
Desirable Overweight Obese (Class I) (Class II) Obese
kg st lb kg st lb kg st lb
0.5 1 40 6 3 75 11 11
1 2 41 6 7 76 12 0
1.5 3 42 6 8 77 12 1
2 4 43 6 11 78 12 5
2.5 6 44 6 13 79 12 6
3 7 45 7 1 80 12 8
3.5 8 46 7 3 81 12 10
4 9 47 7 6 82 12 13
4.5 10 48 7 8 83 13 1
5 11 49 7 10 84 13 3
5.5 12 50 7 13 85 13 6
6 13 51 8 0 86 13 7
52 8 3 87 13 10
10 1 8 53 8 4 88 13 11
15 2 6 54 8 7 89 14 0
20 3 1 55 8 10 90 14 3
21 3 4 56 8 11 91 14 4
22 3 7 57 9 0 92 14 7
23 3 8 58 9 1 93 14 8
24 3 11 59 9 4 94 14 11
25 3 13 60 9 6 95 14 13
26 4 1 61 9 8 96 15 1
27 4 3 62 9 11 97 15 4
28 4 6 63 9 13 98 15 6
29 4 8 64 10 1 99 15 8
30 4 10 65 10 3 100 15 10
31 4 13 66 10 6 101 15 13
32 5 0 67 10 7 102 16 1
33 5 3 68 10 10 103 16 3
34 5 6 69 10 13 104 16 6
35 5 7 70 11 0 105 16 7
36 5 10 71 11 3 106 16 10
37 5 11 72 11 4 107 16 11
38 6 0 73 11 7 108 17 0
39 6 1 74 11 8 109 17 3
Adapted with permission from Webster-Gandy J, Madden A, and Holdsworth M (2006).
Oxford Handbook of Nutrition. Oxford University Press: Oxford
kg st lb kg st lb kg st lb
110 17 5 141 22 3 172 27 1
111 17 7 142 22 5 173 27 3
112 17 8 143 22 7 174 27 6
113 17 11 144 22 10 175 27 8
114 17 13 145 22 11 176 27 10
115 18 1 146 23 0 177 27 12
116 18 5 147 23 1 178 28 0
117 18 6 148 23 5 179 28 3
118 18 8 149 23 6 180 28 5
119 18 10 150 23 8 181 28 7
120 18 13 151 23 11 182 28 9
121 19 0 152 23 13 183 28 11
122 19 3 153 24 1 184 28 14
123 19 6 154 24 3 185 29 2
124 19 7 155 24 6 186 29 4
125 19 10 156 24 7 187 29 6
126 19 11 157 24 10 188 29 8
127 20 0 158 25 13 189 29 11
128 20 1 159 28 0 190 29 13
129 20 5 160 25 3 191 30 1
130 20 7 161 25 5 192 30 3
131 20 8 162 25 7 193 30 5
132 20 11 163 25 9 194 30 8
133 20 13 164 25 12 195 30 10
134 21 1 165 25 14 196 30 12
135 21 3 166 26 2 197 31 0
136 21 6 167 26 4 198 31 3
137 21 8 168 26 6 199 31 5
138 21 10 169 26 9 200 31 7
139 21 13 170 26 11
140 22 0 171 26 13
887
Index
A adrenal disorders
Addison’s disease
MEN-1 587
paraganglioma
abetalipoproteinaemia 835 266–7, 445 284–7, 288–91
acarbose 716, 743 adrenal insufficiency see phaeochromocytoma see
achondroplasia 530 adrenal insufficiency phaeochromocytoma
acipimox 842–3 apparent mineralocorti- staging 253
dose 845 coid excess (AME) 248 adrenal venous sampling 233
acne, PCOS 317–18 autoimmune hyperaldosteronism 244
acromegaly 136, 647 adrenalitis 268–9 adrenalectomy 168, 256, 257
associations 150 autoimmune polyglandular bilateral 611
causes 149, 150 syndrome 270, 271 adrenaline 878
clinical features 151 Cushing’s disease adrenarche, premature 541
and colonic polyps 155–6 250–1, 252–4 adrenocorticotrophic hor-
definition 148 HIV/AIDS 654 mone see ACTH
epidemiology 148 hyperaldosteronism see adrenoleukodystrophy 262
investigations 152–3 hyperaldosteronism age and ageing 613–26
MEN-1 587 hyperplasia see adrenal adrenal function 622
mortality 157 hyperplasia bone disease 616
pregnancy 441 liver disease 659 endocrinology 614–15
treatment 154–7, 155 pregnancy 444 gonadal function 620–1
definition of cure 154 Addison’s disease 445 growth hormone 618
radiotherapy 154 adrenal insufficiency IGF-1 618
surgery 154 277, 445 thyroid disorders 624–6
ACTH 109, 110 CAH 324, 325, 446 Albright’s hereditary
CAH 322 phaeochromocytoma osteodystrophy 476
contraindication in phaeo- 447, 448 albuminuria
chromocytoma 295 renal disease 661 diabetic nephropathy 786
critical illness 662, 663 tumours see adrenal false positive 785
Cushing’s disease 163, tumours alcohol, and diabetes
164, 165 adrenal function 878 mellitus 726
deficiency 123, 265 adrenal hormones 230 aldosterone 229
ectopic ACTH syndrome biosynthesis 321 deficiency see
249, 652–3 older patients 622 hypoaldosteronism
pregnancy 436 see also individual excess see
resistance 664–5 hormones hyperaldosteronism
response to 272 adrenal hyperplasia plasma 878
serum 272, 879 bilateral 239, 254 production 230
ACTH stimulation test congenital see congenital urinary 882
117, 322 adrenal hyperplasia aldosterone-producing
Addison’s disease 266–7 macronodular 653 carcinoma 237
emergency pack 278 adrenal hypoplasia congen- aldosteronoma see Conn’s
pregnancy 445 ita 262, 363 syndrome
see also adrenal X-linked 368 alendronic acid 497
insufficiency adrenal insufficiency alfalfa (Medicago sativa) 678
adolescents 262, 264–5 alkaline phosphatase
diabetes mellitus 762–3 acute 273, 274 as bone turnover
management 764–5 cause 273 marker 452
transition to adult HIV/AIDS 654 thyrotoxicosis 24
services 766–7 investigations 272–4 alpha fetoprotein 881
see also children pregnancy 277 alpha-glucosidase inhibitors
adrenals 227–296 treatment 274, 276–8 715, 716
anatomy 228 adrenal tumours alpha-lipoic acid 677
imaging 232–3 adenoma 252 alprostadil 392
older patients 622 androgen-secreting 328 Alzheimer’s disease, HRT 349
physiology 230 carcinoma 237, 241, 252 ambiguous genitalia 545
pregnancy 444 incidentaloma 282–3 sex assignment 548
888 INDEX
C hyperlipidaemia 824–5
risk factors 825, 826–7
chlorpromazine 295
cholesterol absorption
C-peptide 880 testosterone replacement blocker 843
C-peptide suppression therapy 380 chromium 676
test 632 Carney complex 150, chromogranin A 880
cabergoline 144, 204 250, 583 chromogranin B 880
and cardiac cassia cinnamon chronic fibrosing thyroiditis
valvulopathy 145 (Cinnamomum see Riedel’s thyroiditis
pregnancy 437 aromaticum) 676 Chvostek’s sign 474
café-au-lait spots 540, 576 cataracts, diabetic eye chylomicrons 824
CAH see congenital adrenal disease 779 ciprofibrate 845
hyperplasia catecholamines, hypersecre- citalopram 347
calcitonin 458 tion 288, 289 clivus chordoma 190
complications of cerebral salt wasting 198 clomifene citrate 317,
therapy 502 cerebrovascular disease, 386, 410–11
normal values 880 HRT 348 clomifene test 119, 120
osteoporosis 497 cervical cancer, COCP 358 clonidine 347
calcitriol 497 Charcot foot 808 clonidine suppression test
renal disease 660 CHARGE syndrome 367 290, 291
calcium 451 chemotherapy sensitivity and
circulating 451 endocrine effects 640 specificity 289
serum levels 152, 456 hypogonadism coagulation factors, in
hormonal regulation 458 induced by 373 diabetes mellitus 811
hypercalcaemia see neuroendocrine cocaine 295
hypercalcaemia neoplasias 559 coeliac disease 530
hypocalcaemia see children 513–52 colesevelam 845
hypocalcaemia adrenal disorders, colestipol hydrochloride 845
metabolism 449 Cushing’s disease 173 colestyramine 845
roles of 451 CAH 550–2 collagen crosslinks 452
supplements 502, 609 coeliac disease 530 colonic polyps, and
osteoporosis 680 diabetes mellitus 762–3 acromegaly 155–6
urinary 457, 882 management 764–5 colorectal cancer, HRT
see also bone puberty 766–7 346
calcium/creatinine excretion endocrine effects of combined oral contra-
ratio 457 cancer 642–5 ceptive pill (COCP)
cancer growth 514–17 315, 356–9
childhood, endo- constitutional benefits 356
crine effects in delay 520–1 contraindications 357
survivors 642–5 GH deficiency 522, 524 arterial thrombosis 358
endocrine effects 640 GH resistance 528 venous thromboembo-
see also individual systems rapid 534–5 lism 358
and tumours short stature 518–19 practical issues 359
cannabinoids 677 tall stature 534–5 preparations 357
capecitabine 559 intrauterine growth side effects 356
capsaicin 799 restriction 531 complementary and alterna-
carbimazole 30 puberty 536–7, 537 tive therapy 674
dose 30 delayed 520–1 DHEA 682
side effects 31 delayed/absent diabetes mellitus 676–7
treatment regimen 32 542–3, 543 iodine supplements 682
carbohydrate absorption precocious 538–41 menopause 678–9
inhibitors 677 sexual development osteoporosis 680–1
carcinoembryonic disorders 546–8 computed
antigen 881 sexual differentiation 544 tomography see CT
carcinoid crisis 560 ambiguous genitalia 545 congenital adrenal
carcinoid heart disease skeletal dysplasias 530 hyperplasia
560 small for gestational age 531 adults 320–1, 321
carcinoid tumours, thyroid disorders follow-up 326
MEN-1 587 cancer 102 investigations 322–3
cardiovascular disease hyperthyroidism 48 prognosis 326
diabetes mellitus 810–13 hypothyroidism 7, treatment 324–5
diabetic nephropathy 787 84–5, 529 children 550–2
HRT 349 Turner’s syndrome 532–3 enzyme deficiencies 323
890 INDEX
maturity onset of the foot examination 798–9 see also individual hor-
young (MODY) 691–2 mononeuropathies 795 mones and conditions
mitochondrial 692–3 peripheral sensorimotor Edmonton Obesity Staging
neonatal 692, 763 neuropathy 794 System 849
perioperative management proximal motor eflornithine 316
742, 743, 744 neuropathy 796 electrolyte balance, older
pregnancy 752–5 treatment 799 patients 614
fetal monitoring 754 diabetic retinopathy 768–9 emergency contraception 360
gestational background 770 empty sella syndrome 123
diabetes 756–8 classification 769 endometrial cancer
glycaemic control 754 clinical and histological HRT 348
MODY 692 features 770–2 and PCOS 311
pre-conception pre-proliferative 770–1 endometriosis
management 753 proliferative 771–2 HRT 349
renal disease see diabetic risk factors 776 and infertility 406
nephropathy treatment 788–90 ependymoma 190
secondary 763 blood pressure 788 ephedrine 295
sick day rules 700 cardiovascular risk eplerenone 246
stroke patients 748 factors 788–9 erectile dysfunction 388
type 1 686 diet 789 in autonomic
children and adolescents glycaemia 788 neuropathy 801–2
762, 764–5 referral 789, 790 causes 391
genetics 690 diarrhoea, autonomic 802 definition 388
immune modulation diet evaluation 390–1
therapy 710 diabetes mellitus 713 in older age 621
insulin therapy see diabetic retinopathy 789 treatment 392–4
insulin hyperlipidaemia 838–9 etidronate 497
islet cell obesity 859–60 etomidate 171
transplantation 711 differential diagno- everolimus 559
pancreas sis of endocrine exenatide 716, 717
transplantation 712 disorders 666–9 exercise 672–3
sport and exercise 724–5 dihydrotestosterone 878 bone health 672
type 2 686, 713 dipeptidyl peptidase-4 doping 673
bariatric surgery inhibitors 715, 717 female athlete triad 673
719, 864–5 dong quai (Angelica hypothalamo-pituitary
children and sinensis) 679 axis 672
adolescents 762 dopamine agonists 205 eye examination in diabetes
dietary/lifestyle acromegaly 156 mellitus 774–5
advice 713 pituitary tumours 144, ezetimibe 843
genetics 690 204 dose 845
insulin therapy see insulin resistance 145, 146
oral hypoglycaemics side effects 204
714–17, 715 Down’s syndrome 150 F
and PCOS 311 driving, diabetes familial combined
sport and exercise 725 mellitus 722–3 hyperlipidaemia 832
diabetic amyotrophy 796 drug-induced conditions, familial dysbetalipopro-
diabetic foot 804–5 thyroiditis 73 teinaemia 833
clinical features 805 DSD see sexual differention familial hypercholesterolaemia
epidemiology 805 disorders 830–1
foot examination 798–9 dual-energy absorptiometry familial
protection 739 (DXA) 454 hypertriglyceridaemia 832
risk factors 804 duloxetine 799 familial hypocalciuric
treatment 806–7 dyslipidaemias see hypercalcaemia 470,
Wagner’s classification 805 hyperlipidaemias 471–2, 598
diabetic nephropathy 782–3 familial isolated
definition 782, 783 hyperparathyroidism 596
diagnosis 784–7 E familial mixed
relative risk 783 eating disorders 222 dyslipidaemias 835
diabetic neuropathy ectopic hormone Fanconi syndrome 485
792–3, 793 production 646–7 fasting (Ramadan), diabetes
autonomic ACTH 249, 652–3 mellitus 729
neuropathy 800–3 growth hormone 149 fatigue 667
892 INDEX
causes 75
childhood cancer
sensitivity 698–9
type 2 diabetes 718
K
survivors 643 types of 695, 696 Kallman’s syndrome 362,
congenital 7, 84–5, 529 basal 698 366, 547
fetal 429 quick-acting 698 ketoacidosis 734–6
and hyperlipidaemia 836 insulin growth factor bind- children 765
older patients 624 ing protein-3 (IGFBP-3), ketoconazole 171, 196
post-partum 432 liver disease 659 ketosis-prone diabetes 688
post-radioiodine 35 insulin growth factor-1 Klinefelter’s syndrome
subclinical 78–9 (IGF-1) 132, 152 361, 370
treatment 80–3, 81, 83 critical illness 663 kudzu (Pueraria lobata) 678
generation test 521
liver disease 659
I normal values 879 L
ibandronic acid 497 pregnancy 436 laboratory investigations
IGF-1 see insulin growth insulin pumps 697, autoimmune
factor-1 704–6, 705 adrenalitis 269
imaging see individual modes children 764 factors influencing868–9
imipramine 295, 799 closed-loop systems 710 thyroid disorders 24
immune response in critical insulin secretagogues see also specific conditions
illness 663 714–15, 743 Langerhans cell
in vitro fertilization (IVF) rapid-acting 715 histiocytosis 192
317, 407 insulin sensitizers 676–7 lanreotide 204
incidentaloma insulin tolerance test Laron syndrome 528
adrenal 282–3 114–15 laser treatment, diabetic eye
pituitary 182–3 insulin-to-carbohydrate disease 778
incretin-based therapies ratio 698 latent autoimmune dia-
716–17, 717 insulinoma 562 betes of adulthood
inferior petrosal sinus sam- biochemical features 633 (LADA) 688
pling 164, 167 MEN-1 587 Laurence-Moon-Biedl
infertility 396–7 prognosis 564 syndrome 364, 367
ART 407 treatment 564 lecithin:cholesterol
definition 396 tumour localization 563 acyltransferase (LCAT)
female 396, 398–400, 400 interferon α, neuroendo- deficiency 835
treatment 406–7, 407 crine neoplasias 559 leptin 855
male 397, 402–3, 403 intermediate density lipo- levothyroxine 81
age-related 621 proteins (IDLs) 824 Leydig cells 298
treatment 408–9 interstitial cells 298 licorice (Glycyrrhiza
ovulation intracytoplasmic sperm glabra) 678
induction 410–13 injection (ICSI) 407 Liddle’s syndrome 258
PCOS 317 intrauterine growth restric- lipids 824–5
inherited endocrine tion (IUGR) 531 measurements 827
syndromes 575–600 intrauterine insemination see also hyperlipidaemia
see also specific syndromes (IUI) 407 lipid-lowering drugs 815–16,
inhibin B 376 123iodine- 840–4, 845
normal values 879 metaiodobenzylguanidine acipimox 842–3
insulin see MIBG aims of treatment 844
children 764 ischaemic heart disease, anion exchange resins 842
dawn phenomenon 699 post-menopausal 342 choice of 844
dose adjustments 699 islet cell transplantation 711 cholesterol absorption
in hyperglycaemia 740–1 isoflavones 680 blocker 843
hyperinsulinaemia 310 isotope bone scanning 453 fibrates 841–2
in hyperosmolar hypergly- isotretinoin 318 nicotinic acid 842–3
caemic state 739 omega-3 fatty acids 843
in ketoacidosis 736 statins 815, 840–1, 841
normal values 880 J lipodystrophy 836
perioperative jet lag 224 HIV/AIDS-related 657
management 744 Jod-Basedow lipoma, MEN-1 587
in pregnancy 754 phenomenon 86 lipoprotein A, elevated 834
gestational diabetes 757 Joint British Society lipoprotein lipase
regimens 695–7 Coronary Risk deficiency 833
resistance 663, 693, 811 Prevention Score 826 liquorice ingestion 249
896 INDEX